MXPA01011052A - Therapeutic applications of pro-apoptotic benzodiazepines. - Google Patents

Therapeutic applications of pro-apoptotic benzodiazepines.

Info

Publication number
MXPA01011052A
MXPA01011052A MXPA01011052A MXPA01011052A MXPA01011052A MX PA01011052 A MXPA01011052 A MX PA01011052A MX PA01011052 A MXPA01011052 A MX PA01011052A MX PA01011052 A MXPA01011052 A MX PA01011052A MX PA01011052 A MXPA01011052 A MX PA01011052A
Authority
MX
Mexico
Prior art keywords
aryl
aliphatic
heterocyclic
benzodiazepine
carbons
Prior art date
Application number
MXPA01011052A
Other languages
Spanish (es)
Inventor
Gary D Glick
Original Assignee
Univ Michigan
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Univ Michigan filed Critical Univ Michigan
Publication of MXPA01011052A publication Critical patent/MXPA01011052A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D243/00Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms
    • C07D243/06Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4
    • C07D243/10Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D243/141,4-Benzodiazepines; Hydrogenated 1,4-benzodiazepines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D243/00Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms
    • C07D243/06Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4
    • C07D243/10Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D243/141,4-Benzodiazepines; Hydrogenated 1,4-benzodiazepines
    • C07D243/161,4-Benzodiazepines; Hydrogenated 1,4-benzodiazepines substituted in position 5 by aryl radicals
    • C07D243/181,4-Benzodiazepines; Hydrogenated 1,4-benzodiazepines substituted in position 5 by aryl radicals substituted in position 2 by nitrogen, oxygen or sulfur atoms
    • C07D243/24Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Pulmonology (AREA)
  • Molecular Biology (AREA)
  • Transplantation (AREA)
  • Dermatology (AREA)
  • Urology & Nephrology (AREA)
  • Pain & Pain Management (AREA)
  • Heart & Thoracic Surgery (AREA)
  • AIDS & HIV (AREA)
  • Biomedical Technology (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Biotechnology (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Neurology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Benzodiazepine compounds, and methods for using those compounds are provided. Some of the benzodiazepine compounds include 1,4-benzodiazepine-2-one and 1.4-benzodiazepine-2,5-dione compounds of structures (I) or (II): wherein R1, R2, R3 and R4 are as defined. The invention also includes enantiomers, pharmaceutically acceptable salts, prodrugs or derivatives of the benzodiazepine compounds. Any one or more of these benzodiazepine compounds can be used to treat a variety of dysregulatory disorders related to cellular death. Such disorders include autoimmune disorders, inflammatory conditions, hyperproliferative conditions, viral infections, and atherosclerosis. In addition, the above compounds can be used to prepare medicaments to treat the above-described dysregulatory disorders. The benzodiazepines can also be used in drugs screening assays and other diagnostic methods.

Description

fc THERAPEUTIC APPLICATIONS OF BENZODIAZEPINAS PRO-APOCTOT CAS DESCRIPTION OF THE INVENTION The present description claims the priority of US Provisional Applications Nos. 60 / 191,855, 5 filed March 24, 2000; 60 / 131,761, filed on April 30, 1999; and 60 / 165,511, filed on November 15, 1999, the contents of which are incorporated herein by reference. The present invention is in the field of 10 medicinal chemistry and relates to other areas such as pharmacology, biochemistry and organic chemistry. In particular, novel chemical compounds are provided, and their therapeutic uses. Multicellular organisms exert control 15 precise about the cell number. The normal structure and function of tissues depends on the maintenance of appropriate cell numbers. A balance between cell proliferation and cell death (White, E. (1996) • Genes Dev. 10: 1-15) achieves this homeostasis. 20 Cell death occurs in almost every type of vertebrate cell through necrosis or through a suicidal form of cell death, known as apoptosis. Apoptosis is activated by a variety of extracellular and intracellular signals that employ a mechanism of 25 common, genetically programmed death (Wyllie, A.H. (1995) Curr. Opin. Gen. Dev. 5: 97-104). As a regulatory process, apoptosis influences diverse biological phenomena including the development of neural architecture, the capacity of the • immune system to discriminate between itself and another, and the cancellation of redundant, damaged or infected cells. It becomes evident that many diseases are associated with dysregulation of the cell death process. The experimental models have established a cause and effect relationship between the derangement in the mechanism that regulates 10 apoptosis or necrosis and the pathogenicity of several • neoplastic, autoimmune and viral diseases (Thompson, C.B. (1995) Science 267: 1456-1462). A well-defined example is the high, aberrant level expression effect of bcl-2 in lymphoma development. The bcl-2 oncogene was identified 15 originally as the genetic element located at the t chromosomal translocation breakpoint (14:18) present in many follicular B-cell lymphomas (Korsmeyer, S.J. (1992) Blood 359: 554-556). Since that discovery, it has been • Conveniently established that the bcl-2 gene product 20 inhibits apoptosis induced by a variety of stimuli and that its potential oncogenic stems from its ability to derail apoptosis (Sentman, CL et al. (1994) Cell 64: 878-888; and McDonnell, TJ and Korsmeyer, SJ (1991) Nauret 349: 254-256). 25 Failed or reduced apoptosis is associated with the development of human autoimmune proliferative lymph syndrome as well as mouse models of this disease. MR-lpr or gdl mice develop lymphadenopathy, splenomegaly, nephritis and arthritis; it also produces 5 large quantities of autoantibodies (Cohen, P.L. and Eisenberg, R.A. (1991) Annu, Rev. Immunol., 9: 243-269). These mice carry loss of function mutations in the genes encoding FAS and Fas ligand, respectively (Adachi, M. et al. (1993) Proc. Na ti.Acad.10 Sci. USA 90: 1756-1760); Takakashi, T. et al. (1994) Cell • 76: 969-976). FAS, a cell surface receptor expressed at all sites, normally generates an apoptotic response by binding to the Fas ligand (Itoh, N. et al. (1991) Cell 66: 233-243). In mice that carry these losses 15 of function mutations, disruption of FAS signaling provides T cells resistant to peripheral suppression by apoptosis (Russell, H. et al, Proc.Na.I.Acid.Sci.USA 90: 4409-4413). The inappropriate survival of • these cells result in a pathological accumulation of 20 T and B cells evidenced by similar neoplastic growth of lymphoid tissues and high-level autoantibody production. In humans, the autoimmune lymphoproliferative syndrome shares similarities with the mouse phenotype including lymphadenopathy, splenomegaly, manifestations 25 autoantibody and autoimmune. Patients with this "" - SSS disease as well carry mutation in the FAS gene (Nagata, S. (1998) J. Hum. Genet. 43: 2-8). The benzodiazepine compounds have been known • traditionally to bind to benzodiazepine receptors in the central nervous system (CNS) and have thus been used to treat various CNS disorders including anxiety and epilepsy. More recently, peripheral benzodiazepine receptors have also been identified, whose receptors may incidentally 10 also be present at the CNS. Benzodiazepines and related structures have pro-apoptotic and cytotoxic properties useful in the treatment of cell growth transformed into tissue culture. There is a therapeutic potential for this class of agents against cancer and other 15 neoplastic diseases. Two specific examples shown are neuroblastoma and ovarian cancer. Neuroblastoma is the most common extracranial solid tumor found in children. Modern treatments, which include chemotherapy, radiation therapy and surgery, do not 20 have significantly reduced the mortality of metastatic neuroblastoma. Novel therapies are needed to improve the survival of children with this disease. It was shown that the benzodiazepine compounds are capable of delaying the growth of these tumor cells. See, 25 Sugimoto, T. et al. (1984) J. Na ti. Cancer Inst. 73: 51-57; Schwab, M. et al. (September 15, 1993) Na ture 305.245-248; and Dive, C. and Wyllie, A.H. (1993) Apóptosis and Cancer Chemotherapy. Oxford Blackwell, pp. 21-56. • Ovarian cancer is difficult to treat because of 5 the chemoresistance shown by the patient to standard chemotherapy drugs. Treatment failures are usually attributed to the emergence of cells resistant to chemotherapy. It was shown that benzodiazepine compounds are able to eliminate cancer cells 10 ovaries that are chemoresistant. See, Pestell, K.E. et • al. (1998) In t. J. Cancer 77 (6): 913-918; Beale, P.J. et al. (2000) Br. J. Cancer 82 (2): 436-440; Ozols, R.F. (Feb. 1999) Semin. Oncol. 26 (1 Suppl.2): 84-89; Liu, J.R. et al. (Sep. 1998) Gynecol. Oncol. 70 (3): 398-403; Chumakov, A.M. et al. 15 (Nov. 1993) Oncogene 8 (11): 3005-3011; and Raynaud, F.I. et al. (Aug. 1996) Br. J. Cancer 74 (3): 380-386. Several benzodiazepine analogs have been reported as analgesic and anti-inflammatory agents. See, for example, U.S. Patent Nos. 4,076,823, 4,110,337, 4,495,101, 4,751, 223 and 5,776, 9 6. U.S. Patents Nos. 5,324,726 and 5,597,915 discloses that some benzodiazepines are cholecystokinin and gastrin antagonists and thus may be useful for treating certain gastrointestinal disorders. 25 Certain benzodiazepines have also been explored i.ia.j-. ? g | á £ ^? j »| tt? as human neutrophil elastase inhibitors and thus potentially useful for treating human neutrophil elastase mediated conditions such as myocardial ischemia, septic shock syndrome, among others. See U.S. Patent No. 5,861,380. U.S. Patent No. 5,041,438 reports that certain benzodiazepines may be useful as anti-retroviral agents. However, as will be apparent from the following description, the present invention provides novel methods and compositions that are different from the methods and compositions described above. This invention provides methods for treating a condition associated with dysregulation of the cell death process in a subject, which comprises administering to the subject an effective amount of a benzodiazepine compound. In one aspect, the benzodiazepines of the class are identified by their inability to bind to a central benzodiazepine receptor or with low affinity to a peripheral benzodiazepine receptor. In a further aspect, the benzodiazepines of the class are identified as having the ability to induce cell death under low serum condition as defined infra. In a further aspect, the benzodiazepine classes are identified by having both of these characteristics observed above. In yet another aspect of this invention, chronic inflammatory conditions are excluded ^ ^^ t ^^ j ¿&¿¿¿* iM specifically from the group of conditions that can be treated by any class of compounds specific to the class of benzodiazepines identified herein. • In one aspect, the benzodiazepine compounds have the structure: or its enantiomer, wherein, R_ is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, Wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) -R7 or -R6-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, ^ ^ te ^ St ^^^^ and jS i &j? hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heteroaryl; • or a pharmaceutically acceptable salt, prodrug or derivative thereof. Cell death can be induced by necrosis, apoptosis, or regulation of the FAS trajectory. Conditions associated with dysregulation of a cell death process include, but are not limited to: 10 autoimmune diseases such as systemic lupus erythematosus, arthritis # Rumatoid, Sjögren's syndrome, graft-versus-host disease, and myasthenia gravis; chronic inflammatory conditions such as psoriasis, asthma, and Crohn's disease; hyperproliferative disorders or neoplasms such as 15 lymphomas of a B cell or a T cell; and other conditions such as osteoarthritis and atherosclerosis. The methods are also provided for using the benzodiazepine compounds to treat the conditions associated with the • dysregulation of cell death, where the condition 20 induced by a viral infection. In addition, in some aspects, methods for treating a viral infection are provided using the benzodiazepines of the present invention. The methods are also provided to co-administer one or more additional agents with the 25 benzodiazepines of the present invention, wherein such additional agents may include antineoplastic agents, immunosuppressants, anti-inflammatory agents, antiviral agents, or radiation. • Cell death to be achieved by the methods and compositions of this invention involves the cell or cells present in a tissue that are: autoimmune or affected by an autoimmune disorder; inflammatory or affected by inflammation; hyperproliferative; virally infected; atherosclerotic or osteoarthritic. 10 Testing and diagnostic methods are provided • also to identify agents useful for treating a condition associated with dysregulation of the cell death process in a subject wherein the ability of a potential candidate agent to induce cell death is assayed 15 by contacting the dysregulated cell with a benzodiazepine compound. The test includes keeping the cell adequate or preferably having little serum. The methods are also presented to prepare medications to treat a condition associated with Disregulation of the cell death process in a subject, wherein the conditions, the affected cells or tissue and the benzodiazepine compounds are described as in the above. The invention also provides novel 1,4-benzodiazepine compounds having the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is -NHC (= 0) -R5, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) -R7 or -R6-C (0 =) -NH-R7, wherein R6 is a aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. These various methods, uses, and compositions are m ^ mn ^ j ^ further describe in more detail in the following. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows a general synthetic scheme • for solid phase synthesis of 1, 4-benzodiazepine 5 2 -one compounds of the present invention. Figure 2 shows a general synthetic scheme for solid phase synthesis of compounds 1, -benodiazepine-2,5-dione of the present invention. Figure 3 represents the disease progression analysis for MRL-lpr mice treated according to the # methods described here (solid line) when compared to controls (dotted line). The percentage of disease-free animals (y axis) is plotted over time (e e x). Figures 4A-4C illustrate the swelling of the foot bearing in MRL-lpr mice treated according to the methods described herein (Figure 4A) when compared to the controls (Figure 4B). Figure 4C is a graphical analysis. 20 Figure 5 shows the structure of Compound 1 (Bz-423). Figures 6A-6B are graphs illustrating the in vitro treatment of NZB / W C splenocytes. Figure 6A shows dose response lymphotoxicity of Compound 1. 25 The relative viability expressed by viable cells after treatment as a percentage of viable control cells to allow meaningful comparison between multiple experiments. Figure 6B shows a comparison of Compound 1 to other benzodiazepine receptor ligands and protection against Compound 1 removed by CsA. The compounds are compared at 10 and 20 μM, 10 μM show results. At 20 μM, only Compound 1 demonstrated any cytotoxic effect (data not shown). Figure 7 illustrates the in vitro analysis of cell populations. Experimental conditions are identical to Figure 6. The solid bars represent the percentage of viable cells stained with B220 (B cells) after treatment with the indicated agent. The underlined bars represent cells stained with Thyl .2 (T cells). Figure 8 is a series of photographs showing representative glomerulia of NZB / W mice. Deposition of panel 1 - H &E, panel 2 - IgG, and panel 3 - complement C3 deposition. All sections are photographed at 400X. Figure 9 shows that treatment with Compound 1 prevents the development of autoimmune glomerulonephritis. The kidney sections stained H & E were analyzed by grade of nephritis using a scale of 0-4 +. The - ^ fc, .... ** ^. ---------- solid bars represent mice with disease (> 2+) at sacrifice time, while underlined bars represent healthy mice (< 2+); p < 0.003. Figure 10 is a photograph of representative spleen sections of NZB / W mice. H & E sections of low power - Panel 1 (50X) indicating a decrease in lymphoid content in treatment of mice. Sections of mean power - Panel 2 (200X) indicating increase in numbers and clustering of positive TUNNEL cells in the treatment of mice. High power sections - Panel 3 (400X) co-stained for positive TUNNEL cells (green) and B220 (red) indicating increased numbers of positive B220 + TUNNEL cells in the treatment of mice. Figure 11 is a bar graph illustrating the efficacy of using benzodiazepine to remove D2 neuroblastoma cells in vitro. Figure 12 is a graph showing that cells 2B1 and SKOV3 are resistant to CDDP. Figure 13 is a graph showing that ovarian cancer cells are eliminated by benzodiazepine application in vitro. Throughout this description, several publications, patents and published patent specifications reference by an identified indication. The descriptions of these publications, ^ | ^ imtmmt í É ai? Patent and published patent specifications are incorporated herein by reference in the present description to more fully describe the state of the art to which this invention pertains. A. General Techniques The practice of the present invention will be employed, unless otherwise indicated, conventional techniques of organic chemistry, pharmacology, molecular biology (including recombinant techniques), cell biology, biochemistry, and immunology, which are within the scope of the technique. Such techniques are fully explained in the literature, such as, "MOLECULAR CLONING: A LABORATORY MANUAL" Second Edition (Sambrook et al., 1989); "OLIGONUCLEOTIDE SYNTHESIS" (M.J. Gait, ed., 1984); "ANIMAL CELL CULTURE" (R. Freshney, ed., 1987); the series "METHODS IN ENZYMOLOGY" (Academic Press Inc.); "HANDBOOK OF EXPERIMENTAL INMUNNOLOGY" (D.M. Weir &C.C. Blackwell, ed.); "GENE TRANSFER VECTORS FOR MAMMALIAN CELLS" (J.M. Miller &M.P. Calos, eds., 1987); "CURRENT PROTOCOLS IN MOLECULAR BIOLOGY" (F.M. Ausubel et al., Eds., 1987, and periodic updates); "PCR: THE POLYMERASE CHAIN REACTION" (Mullis et al., Eds., 1994); and "CURRENT PROTOCOLS IN INMUNOLOGY" (J. E. Coligan et al., eds., 1991). B. Definitions As used herein, certain terms may il-tt_iil_¡_tt ___ l¡_li__i have the following defined meanings. As used in the specification and claims, the singular form of "a", "one" and "the" include plural references unless the context clearly dictates otherwise. For example, the term "a cell" includes a plurality of cells, including mixtures thereof. Similarly, the use of "a compound" for treatment or preparation of medicaments as described herein contemplates using one or more compounds of this invention for such treatment or preparation unless the context clearly dictates otherwise. As used herein, the term "comprising" is intended to mean that the compositions and methods include the elements recited again, but does not exclude others. "That consists essentially of" when used to define compositions and methods, which has means that exclude other elements of any meaning essential to the combination. Thus, a composition consisting essentially of the elements as defined herein could not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives, and the like. "Consisting of" has means that exclude more than trace elements from other ingredient steps and substantial method for administering the compositions of this invention. The modalities defined for each of these transition terms are within the scope of this invention. • A "benzodiazepine" refers to a non-aromatic heterocyclic seven-membered ring fused to a phenyl ring wherein the seven-membered ring has two nitrogen atoms as part of the heterocyclic ring. In some aspects, the two nitrogen atoms are in 1 and 4 positions, as shown in the general structure 10 next.
The benzodiazepine may be substituted with a keto group (typically at position 2), or with two keto groups (each at positions 2- and 5). When benzodiazepine has two keto groups (ie, each in 15 positions 2 and 5), is referred to as benzodiazepine-2, 5-dione. More generally, the benzodiazepine is further substituted either in the six-membered phenyl ring or in the seven membered heterocyclic ring or in both rings by a variety of substituents. These substituents are 20 describe more fully in the following. The term "dysregulation of the death process ttl-tf-Mll¡ _ - ___ Í_M_ÍÍÉ Aséj j ^^^ cellular "is intended to cover any aberration in the ability of (or predisposition of) a cell to undergo cell death through either necrosis or apoptosis. • dysregulation of cell death is associated with, or induced by, a variety of conditions, including for example, autoimmune disorders (e.g., systemic lupus erythematosus, rheumatoid arthritis, graft-versus-host disease, myasthenia gravis, Sjogren's syndrome, etc.) .), chronic inflammatory conditions (eg, psoriasis, 10 asthma and Crohn's disease), hyperoproliferative disorders (e.g., tumors, B cell lymphomas, T cell lymphomas, etc.), viral infections (e.g., herpes, papilloma, HIV), and other conditions such as osteoarthritis and atherosclerosis. 15 It should be noted that when dysregulation is induced by, or associated with, a viral infection, the viral infection may or may not be detected at the time of dysregulation or is observed. That is, viral induced dysregulation can occur even after the disappearance of 20 symptoms of viral infection. A "subject" is a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, apes, humans, farm animals, animals for sports, and pets. 25 An "effective amount" is a sufficient amount ^ • ^ tfj to perform beneficial or desired results. An effective amount may be administered in one or more administrations, applications or doses. An "autoimmune disorder" is any condition in which an organism produces antibodies or immune cells that recognize the body's own molecules, cells or tissues. No limiting example of autoimmune disorders includes rheumatoid arthritis, Sjogren's syndrome, graft-versus-host disease, myasthenia gravis, and systemic lupus erythematosus. A "hyperproliferative disorder" is any condition in which a localized population of cells that proliferate in an animal is not governed by the usual limitation of normal growth. Examples of hyperproliferative disorders include tumors, neoplasms, lymphomas and the like. A neoplasm is to be benign, if it is not subjected to, invasion or metastasis and malignant if it is any of these. A metastatic cell or tissue means that the cell can invade and destroy neighboring body structures. Hyperplasia is in the form of cell proliferation that implies an increase in the cellular number in a tissue or organ, without significant alteration in structure or function. Metaplasia is a form of controlled cell growth in which one type of cellular substituent is completely differentiated by another type of cell .. u.?aiem-1.- ^^ "Afc'1iÉgÍ ||| íIÍÉ_ri differentiated.Metaplasia can occur in cells of epithelial or connective tissue.A typical metaplasia involves some disorderly metaplastic epithelium.A" chronic inflammatory condition " "will mean those conditions that are characterized by a persistent inflammatory response with pathological sequelae.This condition is characterized by infiltration of mononuclear cells, proliferation of fibroblasts and small blood vessels, increased connective tissue, and destruction of tissue." Examples of chronic inflammatory diseases include of Crohn, psoriasis, and asthma Autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus can also result in a chronic inflammatory state "Co-administration" refers to the administration of more than one agent or therapy to a subject. By "sensitizing agent" is meant any agent that increases the sensib of an objective cell or tissue to other therapeutic agents. In the context of the present invention, co-administration of the claimed compounds results in a surprising and unexpected effect, particularly in the treatment of conditions in which apoptosis or necrosis is dysregulated. The compounds described herein also appear to sensitize target cells to the therapeutic actions of other agents. The co-administration may be present or, alternatively, the chemical compounds described herein may be administered in advance of, or following the administration of, or the other agents. The appropriate dose for co-administration can easily be determined by one skilled in the art. When co-administered with another therapeutic agent, both agents can be used at lower doses. Thus, co-administration is especially desirable when the claimed compounds are used to decrease the required dose of known toxic agents. "Toxic" refers to any harmful or harmful effects on a cell or tissue. A "composition" is intended to mean a combination of active agent and another compound or composition, inert (eg, a solid support, a detectable or labeled agent) or active, such as an adjuvant. As used herein, "solid phase support" or "solid support", used interchangeably, is not limited to a specific type of support. Rather, a large number of supports are available and are known to one of ordinary skill in the art. The solid phase supports include silica gels, resins, plastic-derived films, glass beads, cotton, pearls of ^ jfcg ^ i ^^^ j plastic, alumina gels. As used herein, "solid support" also includes matrices that present synthetic antigen, cells, and liposomes. A suitable solid phase support can be selected on the basis of the desired end use and convenience for various protocols. For example, for peptide synthesis, the solid phase support can refer to resins such as polystyrene (eg, PAM resin, obtained from Bachem, Inc., Peninsula Laboratories, etc.), POLYHIPE® resin (obtained from Ammotech, Canada), polyamide resin (obtained from Peninsula Laboratories), polyethylene resin grafted with polyethylene glycol (TentaGel®, Rapp Polymere, Tubingen, Germany) or polydimethylacrylamide resin (obtained from Milligen / Biosearch, California). A "pharmaceutical composition" is intended to include the combination of an active agent with a carrier, inert or active, making the composition suitable for diagnostic or therapeutic use in vi tro, in vivo or ex vivo. As used herein, the term "pharmaceutically acceptable carrier" encompasses any of the standard pharmaceutical carriers, such as phosphate buffered saline, water, and emulsions, such as an oil / water or water / oil emulsion, and several types of wetting agents. The compositions may also include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see Martin, REMINGTON'S PHARMACEUTICAL SCIENCES, 15thEd., Mack Publ. Co., Easton, PA (1975). "Sal, prodrug or pharmaceutically acceptable derivative" as used herein, is related to any pharmaceutically acceptable salt, ester, ether, salt of an ester, solvate, such as ethanolate, or other derivative of a compound of the present invention which , under administration to a receptor, is capable of providing (directly or indirectly in the case of a prodrug) a compound of this invention or an active metabolite or residue thereof. Particularly the favored derivatives and prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a mammal (i.e., by allowing a compound administered orally to be more readily absorbed in the blood) or that improves delivery from the paternal compound to a biological compartment (for example, the brain or lymphatic system). As is known to those of ordinary skill in the art, "salts" of the compounds of the present invention can be derived from inorganic or organic acids and bases. Examples of acids include, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p- sulphonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene-2-sulphonic and benzenesulfonic acids. Other acids, such as oxalic, although not pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts. Examples of bases include alkali metal (e.g., sodium) hydroxides, alkaline earth metal (e.g., magnesium), hydroxide, ammonia, and compounds of the formula NW4 +, wherein W is C? - alkyl. Examples of salts include: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorrate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecyl sulfate, ethanesulfonate, fumarate, flucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide. , hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, pamoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate and undecanoate. Other examples of salts include anions of the compounds of the present invention compounds with a suitable cation such as Na +, NH4 +, and NW4 + (wherein W is an alkyl group of C? _?). For therapeutic use, the salts of the compounds of the present invention will be pharmaceutically acceptable. However, salts of acids and bases that are not pharmaceutically acceptable can also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound. The term "derivative" of a compound as used herein means a chemically modified compound wherein the chemical modification takes place either in a functional group of the compound or in the aromatic ring. Non-limiting examples of 1,4-benzodiazepine derivatives of the present invention may include N-acetyl, N-methyl, N-hydroxy groups in any of the available nitrogens in the compound. Additional derivatives may include those having a trifluoromethyl group on the phenyl ring. C. Treatment Methods The therapeutic potential for using benzodiazepines for their pro-apoptotic and cytotoxic properties is greater. The class of agents is effective in treating cancers and other neoplastic diseases. As noted above, the present invention provides methods for treating conditions that are, in one embodiment, related in that they arise as the result of deregulation of the normal process of cell death in the cells or tissue of a subject.
For the purpose of illustration only, such conditions include, but are not limited to, autoimmune disorders (e.g., systemic lupus erythematosus, • rheumatoid arthritis, graft-versus-host disease, 5 Sjögren's syndrome, and myasthenia gravis); hyperproliferative disorders (such as B-cell and T-cell lymphoma, neuroblastoma, and chronic lymphocytic leukemia); chronic inflammatory conditions (such as psoriasis, asthma, or Crohn's disease); other conditions such as osteoarthritis and atherosclerosis, and those induced by • Viral DNA and / or RNA infections, where the viruses include, but are not limited to, herpes virus, papilloma virus and human immunodeficiency virus (HIV). These disorders are treated by administering an effective amount of the benzodiazepine compounds described herein. The various benzodiazepine compounds are described more fully in the following. These compounds are therapeutically effective in themselves, and have little or no toxic effect when administered in prolonged doses. In addition, as described in detail in the following, the co-administration of these compounds with other agents provides an unexpected synergistic therapeutic benefit. In co-administration methods, the claimed compounds are also useful in reducing harmful side effects of therapeutic agents .uní i.ir.ini-ilitiHÉ iiftlffltif i | i ||, iiM_i_t_t -_-_-_-_- i-_Í_Í_t_t-_i «-MM known by decreasing the amount which will be administered to the subject. The conditions which benefit from the treatment with the compounds described herein appear to divide the common etiology of dysregulation from the cell death process. As described above in the Background, normal apoptosis occurs through several trajectories, with each trajectory having multiple stages. The methods described herein are useful in treating dysregulated apoptosis despite the trajectory or stage in the trajectory where the dysfunction occurs. In one embodiment, the condition is triggered as the result of deregulation of the apoptotic FAS path. Similarly, the compounds are also useful in treating dysregulated necrosis despite the trajectory or stage in the trajectory where the dysfunction occurs. The dysregulation of the cell death process is associated with many conditions. In neoplasms, for example, normal cell death is inhibited, allowing hyperproliferative growth of cells. The aberrant functioning of this process can also result in serious pathologies including autoimmune disorders, viral infections, conditions induced by viral infections, neurodegenerative disease, and the like. The present invention provides methods for treating these and other conditions. tt áté m? Miik? mm Without being limited by a theory, it seems that the effective compounds described herein induce or promote cell death when this process is a defective process. Thus, in addition to treating conditions associated with dysregulation of apoptosis, the compounds of this invention also treat conditions in which no apoptotic defect can exist. For example, in certain viral infections, although no apoptotic defect can exist, cell death can be promoted by inducing necrosis. The condition to be treated is generally determined by noting the presence of symptoms in the subject or noting phenotypic or genotypic changes in the cells of the subject, in particular, the inability of the cell to undergo apoptosis or necrosis. The phenotypic changes associated with the neoplastic state of a cell (a group of characteristics in vi tro associated with a tumorigenic capacity in vivo) include more rounded cell morphology, looser substrate binding, loss of contact inhibition, loss of dependence on anchoring, protease release, increased sugar transport, decreased serum requirement, expression of fetal antigens, etc. See, Luria, et al. , (1978) GENERAL VIROLOGY, 3rd edition, pp. 436-446 (John Wiley &Sons, New York). To "treat" as intended herein, it means to induce cell death (where cell death is either aproptotic or necrotic) in cells or tissue that are causative (primary or distal) of the disorder being treated. For example, in hyperproliferative disorders, the method • treat the disorder by inducing apoptosis of the hyperproliferative cells, such as neoplastic cells. In this modality, the reduction in tumor size or tumor burden is a means to identify that the object of the method has met. In other aspects, "to treat" accompanies restoration of immune function or regulation of immune dysfunction 10, such as an autoimmune disorder and conditions • Chronic inflammatory. In other aspects, the viral concentration is eliminated or reduced in the subject being treated. In other aspects, "to treat" means to lessen the symptoms associated with a particular disease, for example, cachexia in cancer or HIV infection or inflammation in arthritis. In still further aspects, the prophylaxis as well as the therapeutic use of the compounds and methods of this invention are intended. In the embodiment wherein the condition that is treated is an autoimmune disease, the use of the methods described herein will reduce the autoantibody production and lead to a decrease in inflammation and tissue destruction. Thus, the cell that is being treated can be the cell that itself is self-immunogenic or is affected distally by an autoimmune reaction, where it is , s. s; i. It is desirable to induce cell death in such cells or tissues containing such cells. Similarly, in the case of treating inflammatory conditions, the cell being treated may be the inflammatory cell itself or is affected distally by inflammation where it is desirable to induce cell death in such cells or tissues containing such cells and thus reduce inflammation. In a further embodiment, the cell that is treated is a virally infected cell or a cell or 10 tissue that has previously been infected. Successful therapy • induces cell death and therefore a reduction in viral concentration. This result is easily determined by testing the viral concentration or by observing a reduction in the cell number. It should be noted that, as can be inferred 15 from the statements, supra, may be desired to induce cell death even among those cells that do not have any viral residue or other signs of viral infections at the time of treatment due to viral infection • What happens much earlier in time could still cause 20 interruption of cell death to a very subsequent time. Cell death can be assayed as described herein and in the art. The cell lines are maintained under appropriate culture conditions (eg, gas (C02), temperature and medium) and for an appropriate amount 25 of time to reach exponential proliferation without , K n,? T. > ? i. liiíiiriHiili it ii ¡ím * * ^ > A? ^ M ^. ^ ^ dependent density. The cell number and / or availability can be measured using standard techniques, such as trypan blue / heme-cytometry exclusion, or • MTT dye conversion test. Alternatively, the cell can be analyzed for the expression of genes or gene products associated with aberrations in apoptosis or necrosis. It should be understood that the various dysregulatory disorders described above can be treated by any of one or more benzodiazepine compounds, 10 including many alternative and specific modalities • presented in this. These benzodiazepines are described more fully in the following. Antiviral Activity In another embodiment, the compounds of the present The invention may have antiviral activity independent of its efficacy to induce cell death. An aspect or method for inhibiting viral replication and / or propagation comprises contacting the virus with an effective amount of one or more compounds and / or compositions of the invention. 20 present invention. The contact is conducted under suitable conditions to inhibit viral replication and / or propagation. In another aspect, the method comprises preventing viral infection and / or propagation in a cell or tissue by contacting the cell or tissue with an amount Effective of the compounds and / or compositions as defined above. The contact is conducted under suitable conditions so that the viral infection and / or spread is inhibited. The inhibition or prevention of viral infection, replication and / or spread can be measured by assaying by viral concentration, which methods are well known to those skilled in the art. By inhibiting and reducing viral replication and proliferation, viral infectivity is also inhibited and reduced and host cells are treated appropriately for viral infection with the additional benefit that the associated pathologies are also treated. As used herein, the term "suitable conditions" includes conditions in vi tro, ex vivo or in vivo. These terms are well known in the art. The viruses contemplated under the present methods include RNA and / or DNA viruses. By way of example only, such viruses are herpes, without herpes and of retroviral origin. The principal examples of human pathogens of the herpes virus family include herpes simplex virus (HSV) 1,2, and herpes virus cercopithecine 1 (virus B); varicella zoster; Epstein-Barr virus (EBV); Lymphoprotovirus; human herpesvirus 6-8 (HHV6-S); herpes virus associated with kaposi (KHV); simian herpesvirus; and human cytomegalovirus (HCMV). See, for example, Gallant, M ^ J.E. et al., J. Infect. Dist. 166: 1223-1227, (1992). Animal pathogens of herpesviral origin include infectious bovine rhinotracheitis virus, bovine mamilitis virus, and cercopithecine herpes virus (B virus), among others. Human viruses of origin without herpes include influenza A, B and C viruses; parainfluenza virus -1, 2, 3 and 4; adenovirus; reovirus; respiratory syncytial virus; rhinovirus; coxsackie virus; eco virus; rubella virus; hepatitis B virus types B and C (HBV and HCV); and papovavirus. Animal viruses of no herpes origin include pseudorabies virus (PRV, from pig), equine rhinopneumonitis, coital exanthema virus (varicella virus); lymphoprotovirus; Marek's disease virus, Herpesvirus-1 Bovine (BHV-1), virus Pseudorabias herpesvirus (PRV). Viruses of retroviral origin that are contemplated as being treatable by the compounds and compositions of this invention include human immunodeficiency virus (HIV) types 1 and 2 and human lymphotropic viruses 1 and 2 (HTLV-I and II) • D. Methods for Identifying Potential Therapeutic Agents Also provided herein is an assay to identify a potential agent to treat a condition associated with dysregulation of the apoptotic or necrotic path. The method comprises contacting the dysregulated cell, i.e., a cell affected by the disorder (e.g., a tumor cell, when the • condition is hyperproliferative) or an immune cell (a neutrophil, basophil, eosinophil, monocyte, or lymphocyte) when the condition is a chronic inflammatory condition or an autoimmune disorder) with the agent. In a further aspect of this invention, a control cell is further tested with or without a benzodiazepine compound. The compound Benzodiazepine 10 can be a 1,4-benzodiazepine compound • as described herein. The dead cell when compared to the control cell is also observed and compared. To identify these potential therapeutic agents, the appropriate assay conditions (by Example, incubation time, temperature, culture maintenance medium, etc.) can be easily determined by one of those skilled in the art. The serum can be obtained from any commercial source, • for example, fetal bovine serum from Gibco BRL 20 (Gaithersburg, MD). The cells are cultured with the test agent for a sufficient amount of time. Following the appropriate incubation period, cell death can then be assayed by any means described above, for example, by trypan dye exclusion 25 MTT. Thus, novel cytotoxic agents can TÉrtrtiri i piiiiiiii? ? i? i? l ?? ?? i ??? i? i || ____ i > t ----- J | be identified by its ability to induce cell death to dysregulated cells. In addition, comparisons can also be made for cell death in control cells. It has been found that when the cells are maintained under low serum conditions, the cytotoxicity is greatly exacerbated and the incubation time is reduced to approximately 2 hours or less. This is completely an unexpected result since under standard incubation conditions, which employ high serum levels, the required incubation time is often several hours, approaching in some cases to 24 hours or more. "Little serum" as used herein refers to the culture medium containing less than about 10% by volume below or equal to less than about 0.1% (v / v). It should be understood that within this range the concentration is flexible, and any possible subrange is contemplated in increments of approximately 0.1% within this range, for example, less than or equal to approximately any of 0.1, 0.2, 0.3, 0.4, 0.5 , 0.6 / 0.7, 0.8, ... 1.0, ... 1.5, ... 2.0, ... 2.5, ... 3.0, ... 3.5, ... 5%, etc., to approximately 9.9 and approximately 10% serum (% v / v). Thus, in one aspect, the benzodiazepines of this invention induce apoptosis in little serum as defined above. In a further aspect, the benzodiazepines of This invention is further characterized and identified by its loss of ability to bind to a central benzodiazepine receptor or to bind with low affinity to a peripheral benzodiazepine receptor. These compounds can be identified using methods well known in the art. For example, the binding affinity of a benzodiazepine compound for a peripheral benzodiazepine receptor can be determined according to the methodology 10 well established as described in Schomaker, H. et al. • (1983) J. Pharm. Exp Ther. 225: 61-69 and Doble, A. et al. (1987) Bra m Res. Bull. 18: 49-61. Briefly, the method comprises comparing the potency of a benzodiazepine compound with that of a well known high affinity binding agent such as l- (2-chlorophenyl) -N-methyl-N- (-1, methylpropyl) -3 isoquinolcarboxamide (PK11195), wherein the ability of the benzodiazepine compound to displace PK511195 from the peripheral benzodiazepine receptors in a competitive binding assay. In any of the above test methods, the benzodiazepine compound can be detectably labeled. Such detectable labeling includes labeling with an isotope or with a fluorescent portion. Examples of 25 isotope labeling include use of stable isotopes or yÉj__i_. radioactives of one or more atoms in the benzodiazepine molecule. Methods for introducing such detectable labels and for detecting labels are well known in the art. For example, the radioisotope tag can be detected using special instrumentation, including electron spin resonance spectrometers. Stable isotopes can be detected using mass spectrometers, or magnetic resonance spectrometers. The brands 10 fluorescents can be detected using spectrometers • fluorescents. Many of these instruments are commercially available and their operation is within the ordinary scope in the art. The benzodiazepine compounds that can 15 used in the assay and diagnostic methods are described in more detail in the following. It should be understood that all of the compounds described herein, including the many general and specific embodiments, may be used. • in the assay and diagnostic methodology. E. Use of Benzodiazepine Compounds to Prepare Medicaments The benzodiazepine compounds of the present invention are also useful in the preparation of medicaments for treating a variety of conditions 25 associated with the dysregulation of cell death as ^^ j | É | ^ g, | __l ___ * _ ft_ft_l_ _É _ * _ ^^ described above. In addition, the compounds can also be used to prepare drugs for treating other disorders wherein the effectiveness of benzodiazepines is known or predicted. Such disorders may include neurological or neuromuscular disorders. Methods and techniques for preparing medicaments of a compound are well known in the art. Some possible pharmaceutical formulations and delivery routes are detailed in the following. Thus, one skilled in the art will readily appreciate that any one or more of the compounds more fully described in the following, including the many specific embodiments, can be used by applying standard pharmaceutical manufacturing methods to prepare medicaments for treating the many disorders described herein. previously. Such medicaments can be delivered to the subject using delivery methods that are well known in the art in pharmaceutical techniques. F. Compositions and Formulations Although it is possible for the agent to be administered alone, it is preferable to present it as a pharmaceutical formulation comprising at least one active ingredient, as defined above, together with a solid support or alternatively, together with one or more carriers mm imu ^ i ^ ^ ák? m pharmaceutically acceptable for that and optionally other therapeutic agents. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not harmful to the patient. The formulations include those suitable for oral, rectal, nasal, topical (including transdermal, buccal and sublingual), vaginal, parenteral (including subcutaneous, intramuscular, intravenous and intradermal) and pulmonary administration. The formulations may conveniently be presented as a dosage unit and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of presenting the active ingredient in association with the carrier which constitutes one or more adjunct ingredients. In general, the formulations are prepared uniformly and presented intimately in association with the active ingredient with liquid carriers or finely divided solid carriers or both, and then if necessary forming the product. Formulations of the present invention suitable for oral administration may be present as discrete units such as capsules, wafers or tablets, each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or water-in-oil liquid emulsion. The active ingredient may also present a bolus, remedy or paste. A tablet can be made by compression or • molding, optionally with one or more ingredients attached. The compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g., povidone, gelatin, hydroxypropylmethylcellulose), lubricant, diluent 10 inert, conservative, disintegrating (for example, glycolate of • sodium starch, crosslinked povidone, cross-linked sodium carboxymethyl cellulose) active or dispersing surface agent. Molded tablets can be made by molding in a suitable machine a mixture of the powder compound 15 moistened with an inert liquid diluent. The tablets may be optionally coated or classified and may be formulated so as to provide slow or controlled release of the active ingredient herein using, for example, hydroxypropylmethylcellulose in proportions 20 varied to provide the desired release profile. The tablets may optionally be provided with an enteric coating, to provide release in portions of the intestine other than the stomach. The formulations suitable for administration 25 topical in the mouth include dragees comprising the I-_É_I _-______. lliI iiniil | MJli ^ iiÉ _ ^ -_ t_ | ____ g_i__ri active ingredient in a flavor base, usually sucrose and acacia or tragacanth; Pills comprising the active ingredient in an inert base such as gelatin or • glycerin, or sucrose and acacia; and mouth rinses that comprise the active ingredient in a suitable liquid carrier. Pharmaceutical compositions for topical administration according to the present invention can be formulated as an ointment, cream, suspension, lotion, powder, solution, paste, gel, spray, aerosol or oil. Alternatively, a The formulation may comprise a patch or a dressing such as a band or an adhesive plaster impregnated with the active ingredients, and optionally one or more excipients or diluents. If desired, the aqueous phase of the base cream may include, for example, at least about 30% w / w of a polyhydric alcohol, ie, an alcohol having two or more hydroxyl groups such as propylene glycol, butan- 1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol and mixtures thereof. Topical formulations may desirably include a compound that improves the absorption or penetration of the agent through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethylsulfoxide (DMSO) and related analogues.
The oil phase of the emulsions of this invention can be constituted from known ingredients in a known manner. Although this phase can • comprising only one emulsifier (otherwise known as an emulsifier), this desirably comprises a mixture of at least one emulsifier with a fat or an oil or both of a fat and an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as 10 a stabilizer. It is also preferred to include both an oil and a fat. Together, the emulsifier (s) with or without the stabilizer (s) perform the so-called emulsifying wax, and the wax together with the oil and / or grease performs the so-called emulsifying ointment base that 15 forms the oily dispersed phase of the cream formulations. Emulsifiers and emulsion stabilizers suitable for use in the formation of the present invention include Tween 60, Span 80, ketostearyl alcohol, alcohol 20 myristyl, glyceryl monostearate and sodium lauryl sulphate. The choice of oils or greases suitable for the formulation is based on achieving the desired cosmetic properties, since the solubility of the compound is very low 25 active in most oils is likely to be used ^ M? i a u u m ^ ^ J ^ in pharmaceutical emulsion formulations. Thus the cream should preferably be non-greasy, undyed and the washable product with adequate consistency to avoid filtration of tubes or other containers. The straight or branched chain, 5 mono- or dibasic alkyl esters such as di-isoadipate, isocetyl stearate, propylene glycol diacid of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, palmitate 2- ethylhexyl or a mixture of branched chain esters 10 known as Crodamol CAP can be used, the last three • being the preferred esters. These can be used alone or in combination depending on the required properties. Alternatively, high-melting lipids such as white soft paraffin and / or liquid paraffin or other 15 mineral oils can be used. Formulations suitable for topical administration to the eye, also include eye drops, wherein the active ingredient is dissolved or suspended in a carrier • adequate, especially an aqueous solvent for the agent. Formulations for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter or a salicylate. Formulations suitable for vaginal administration can be presented as intrauterine devices, 25 tampons, creams, gels, pastes, foams or formulations of m, iiitttii íu u? It was sprayed containing, in addition to the agent, such carriers as are known in the art to be appropriate. Formulations suitable for nasal administration, wherein the carrier is a solid, include a coarse powder having a particle size, for example, in the range of about 20 to about 500 microns which is administered in the form in which the sniffing is taken, that is, by rapid inhalation through the nasal passage of a dust receiver that helps close the nose. The 10 suitable formulations where the carrier is a liquid • for administration as, for example, nasal spray, nasal drops, or aerosol administration by nebulizer, which includes aqueous or oily solutions of the agent. The formulations suitable for administration Parenteral drugs include sterile aqueous and non-aqueous isotonic injection solutions which may contain antioxidants, buffers, bacteriostats and solutes which give the formulation isotonic with the blood of the recipient. • intended; and sterile aqueous and non-aqueous suspensions that 20 may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to direct the compound to the blood components or one or more organs. The formulations can be presented in unit sealing containers. 25 doses or multiple doses, for example, ampules or bottles, and «" "- * - * - * -» * »» "- | ^ Ugj ^^^^ can be stored in a freeze drying condition (lyophilized) requiring only the addition of the sterile liquid carrier, eg water for injections , immediately before use Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules and tablets of the previously described genus Preferred dosage unit formulations are those containing a daily dose or unit, daily sub-dose, as hereinafter, it is recited in the foregoing, or an appropriate fraction of the mimic, of an agent It should be understood that in addition to the ingredients particularly mentioned above, the formulations of this invention may include other agents conventional in the art having Consideration of the type of formulation in question, for example, those suitable for oral administration may include such additional agents as sweetening agents, thickeners and flavorings. It is also intended that the agents, compositions and methods of this invention be combined with other suitable compositions and therapies. G. Pharmaceutical Supply Various delivery systems are known and can be used to administer a therapeutic agent of the invention, for example, encapsulation in liposomes, ^ f ^ ^^ m¡ß microparticles, microcapsules, receptor-mediated endocytosis (see, for example, Wu and Wu, (1987), J. Biol. Chem. 262: 4429-4432), and the like. Methods of delivery include, but are not limited to, intra-arterial, intramuscular, intravenous, intranasal, and oral routes. In a specific embodiment, it may be desirable to administer the pharmaceutical compositions of the invention locally in the area in need of treatment; this can be achieved for example, and not by way of limitation, local infusion during surgery, 10 by injection, or by means of a catheter. • Agents identified herein as effective for their intended purpose may be administered to subjects or individuals susceptible to or at risk of developing a condition correlated with dysregulation 15 of the apoptotic or necrotic trajectory. When the agent is administered to a subject such as a mouse, a rat or a human patient, the agent can be added to a pharmaceutically acceptable carrier and systemically or topically administered to the subject. To determine the patients that 20 can be treated beneficially, a tissue sample is removed from the patient and the cells are assayed for agent sensitivity. Therapeutic quantities can be determined empirically and will vary with the pathology that is treated, the 25 subject being treated and the efficacy and toxicity of the agent.
If it is given to an animal, the method is useful to further confirm the effectiveness of the agent. An example of an animal model is MLR / MpJ-lpr / lpr ("MLR-lpr") (available from Jackson Laboratories, Bal Harbor, Maine). The MLR-lpr 5 mice developed the systemic autoimmune disease. Alternatively, other animal models can be developed by inducing tumor growth, for example, by inoculating subcutaneously nude mice with about 105 to about 109 hyperproliferative, cancerous or 10 objective as defined herein. When the tumor is • stabilizes, the compounds described herein are administered, for example, by subcutaneous injection around the tumor. Tumor measurements to determine tumor size reduction are done in two dimensions 15 using venier calibrators twice a week. Other animal models can also be used as appropriate. The animal models for the diseases and conditions described above are well known in the art. • technique. See, for example, (1992) Am. J. Pa thol. 36: 875-208 882. The in vivo administration can be effected in one dose, continuously or intermittently through the course of treatment. Methods to determine the most effective means and dosage of administration are well known 25 by those experienced in the art and will vary with the • ^ t? M iimá M composition used for therapy, the purpose of therapy, the target cell that is treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and the standard being selected by the attending physician. The formulations and dosage methods suitable for administering the agents can be readily determined by those skilled in the art. Preferably, the compounds are administered at about 0.01 mg / kg to about 200 mg / kg, more preferably at about 0.1 mg / kg to about 100 mg / kg, even more preferably from about 0.5 mg / kg to about 50 mg / kg. kg. When the compounds described herein are co-administered with another agent (e.g., as sensitizing agents) the effective amount may be less than when the agent is used alone. The pharmaceutical compositions can be administered orally, intranasally, parenterally or by inhalation therapy, and can take the form of tablets, dragees, granules, capsules, pills, ampules, suppositories or in the form of an aerosol. They can also take the form of suspensions, solutions and emulsions of the active ingredient in aqueous or non-aqueous diluents, syrups, granulates or powders. In addition to an agent of the present invention, the pharmaceutical compositions may also contain other pharmaceutically active compounds or a plurality of compounds of the invention. More particularly, an agent of the present invention is also referred to herein as the active ingredient, can be administered by therapy by any suitable route including oral, rectal, nasal, topical (including transdermal, aerosol, buccal and sublingual) vaginal, parental (including subcutaneous, intramuscular, intravenous and intradermal) and pulmonary. It will be appreciated that the The preferred route will vary with the condition and age of the recipient, • and the disease that is treated. Ideally, the agent should be administered to achieve peak concentrations of the active compound at disease sites. This can be achieved, for example, 15 by intravenous injection of the agent, optionally in saline, or administered orally, for example, as a tablet, capsule or syrup containing the active ingredient. Desirable blood levels of the agent can 20 be maintained by a continuous infusion to provide a therapeutic amount of the active ingredient within the disease tissue. The use of the operative combinations is contemplated to provide therapeutic combinations that require a lower total dose of each The antiviral agent component may be required when each individual therapeutic compound or drug is used alone, thereby reducing the adverse effects. H. Co-Administration The present invention also includes methods that involve the co-administration of the compounds described herein with one or more additional active agents. Indeed, it is a further aspect of this invention to provide methods for improving prior art therapies and / or pharmaceutical compositions by co-administering a 10 compound of this invention. In the methods of cof administration, the agents can be administered simultaneously or sequentially. In one embodiment, the compounds described herein are previously administered to the or the other active agents. Pharmaceutical formulations and modes of The administration can be any of those described above. In addition { Thus, the two or more coadministered chemical agents, biological or radiation agents can each be administered using different modes of different formulations. 20 The agent or agents to be co-administered depend on the type of condition being treated. For example, when the condition being treated is hyperproliferation, the additional agent can be a chemotherapeutic or radiation agent. When the condition that is treated is a disorder 25 autoimmune, the additional agent can be an agent immunosuppressant or anti-inflammatory. When the condition being treated is chronic inflammation, the additional agent can be an anti-inflammatory agent. When the condition is # treated is a viral infection or conditions induced by a viral infection, the addition agent can be an antiviral agent. Additional agents to be co-administered, such as anti-cancer, immunosuppressant, anti-inflammatory, and antiviral agents can be any of the agents well known in the art, including those that are currently in clinical use. The type determination • Appropriate and radiation treatment dose is also within the scope in the art or can be determined with relative ease. Currently, the treatment of the various conditions associated with abnormal apoptosis is limited by the following two main factors: (1) the development of drug resistance and (2) the toxicity of the known therapeutic agents. In certain cancers, for example, the • Resistance to chemicals and radiation therapy have been shown to be associated with the inhibition of apoptosis. See, Desoize, B. (1994) An ticancer Res. 14: 221-224. Similarly, some therapeutic agents have dangerous side effects, including nonspecific lymphoxicity, renal and bone marrow toxicity. 25 The methods described herein are directed to tt m m ^ M? hm im both of these problems. Drug resistance, when the dose is increased is required to achieve therapeutic benefit, is overcome by co-administering the compounds described herein with the known agent. The 5 compounds described herein appear to sensitize target cells to known agents and, consequently, fewer of these agents are necessary to achieve a therapeutic benefit. The sensitizing function of the compounds 10 claimed also addresses the problems associated with • the toxic effects of known therapeutics. In some cases where the known agent is toxic, it is desirable to limit the doses administered in all cases, and particularly in those cases where the drug resistance has 15 increased the required dose. When the claimed compounds are co-administered with the known agent, they reduce the required dose which, in turn, reduces the harmful effects. In addition, because the claimed compounds are • by themselves both effective and non-toxic in large In addition to the dose, the co-administration of proportionally more of these compounds than the known toxic therapeutics will achieve the desired effects while minimizing the toxic effects. I. The Benzodiazepine Compounds The compounds of the present invention are benzodiazepine compounds. In some aspects the benzodiazepine compounds have the following structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= -0) -R5 or a portion that participates in hydrogen bonding, wherein R5 is aryl, heterocyclic, -R? -NH-C (= 0) -R7 or -R6 ~ C (= 0) -NH-R- ?, wherein Rβ is an aliphatic linker of 1-6 carbons and R is aliphatic, aryl or heterocyclic, each of R and R4 is independently a hydroxy, alkoxy, halo, amino, lower alkyl-amino It is substituted, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic, or a pharmaceutically acceptable salt, prodrug or derivative thereof. the above structures, Ri is a hydrocarbyl group of 1-20 carbon atoms and 1-20 hydrogens Preferably, Ri has 1-15 carbons, and more preferably has 1-12 carbons Preferably Ri has 1-12 hydrogens, and more preferably, 1-10 hydrogens Thus, R_ may be an aliphatic group or an aryl group The term "aliphatic" represents the groups commonly known as alkyl, alkenyl, alkynyl, alicyclic, the term "aryl" as used in the present represents a single aromatic ring such as a phenyl ring, or two or more aromatic rings which are connected together (eg, bisphenyl) or fused together (eg, naphthalene or anthracene) The aryl group may be optionally substituted with a lower aliphatic group (eg, C_-C, alkenyl, alkynyl, or C3-Ce alkyl alicyclic). Additionally, the aliphatic and aryl groups can be further substituted by one or more functional groups such as -NH2, -NHCOCH3, -OH, lower alkoxy (C1-C4), halo (-F, -Cl, -Br, or -I ). It is preferred that Ri is primarily a non-polar portion.
In the above structures, R 2 may be aliphatic, aryl, -NH 2, -NHC (= 0) R 5, or a portion that participates in hydrogen bonding, wherein R 5 is aryl, heterocyclic, R 6 -NH-C (= 0 ) -R7 or -R6-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is an aliphatic, aryl or heterocyclic linker. The terms "aliphatic" and "aryl" are as defined above. The term "a portion that participates in hydrogen bonding" as used herein represents a group that can accept or donate a proton to form a hydrogen bond accordingly. Some specific non-limiting examples of the portions involved in hydrogen bonding include a fluorine, oxygen containing and nitrogen containing groups that are well known in the art. Some examples of oxygen-containing groups that participate in hydrogen bonding include: hydroxy, lower alkoxy, lower carbonyl, lower carboxyl, lower ethers and phenolic groups. The "lower" qualifier as used herein refers to lower aliphatic groups (C? -C4) to which the respective oxygen-containing functional group is attached. Thus, for example, the term "lower carbonyl" refers to inter alia, formaldehyde, acetaldehyde. Some non-limiting examples of the nitrogen-containing groups involved in bond formation of hydrogen include amino and amido groups. Additionally, groups containing an oxygen atom and a nitrogen atom may also participate in the binding formation • of hydrogen. Examples of such groups include nitro, N-5 hydroxy and nitroso groups. It is also possible that the hydrogen-binding acceptor in the present invention can be the p-electrons of an aromatic ring. However, the hydrogen bonding partners of this invention do not include those groups containing metal atoms such as boron. further • of the hydrogen bonds formed within the scope of the practice of this invention do not include those formed between two hydrogens, known as "dihydrogen bonds". See, Crabtree, R.H. (1998) Science 282: 2000-2001, for further description of such diacid linkages. The term "heterocyclic" represents an aromatic or non-aromatic 3-6 membered ring containing one or more heteroatoms. The heteroatoms may be the same or different from each other. Preferably, at least one of the 20 heteroatoms is nitrogen. Other heteroatoms that may occur in the heterocyclic ring include oxygen and sulfur. Heterocyclic aromatic and non-aromatic rings are well known in the art. Some non-limiting examples of heterocyclic aromatic rings include . at -t-_ ~ -.J. »a-. < ___ a¿_M ^, .. ". , .._ * _ ^ * .. ¡, *, ..- * .. ",. l É |||| i, í, | ^ ^ ^ i pyridine, pyrimidine, indole, purine, quinoline and isoquinoline. Non-limiting examples of non-aromatic heterocyclic compounds include piperidine, piperazine, morpholine, pyrrolidine and pyrazolidin. Examples of oxygen containing heterocyclic rings include, but are not limited to furan, oxirane, 2H-pyran, 4H-pyran, 2H-chromene, and benzofuran. Examples of heterocyclic rings containing sulfur include, but are not limited to, thiophene, benzothiophene, and parathiazine. Examples of nitrogen-containing rings include, but are not limited to, pyrrole, pyrrolidine, pyrazole, pyrazolidin, imidazole, imidazoline, imidazoiidine, pyridine, piperidine, pyrazine, piperazine, pyrimidine, indole, purine, benzimidazole, quinoline, isoquinoline, triazole and triazine. Examples of heterocyclic rings containing two different heteroatoms include, but are not limited to, phenothiazine, morpholine, parathiazine, oxazine, oxazole, thiazine, and thiazole. The heterocyclic ring is further optionally substituted with one or more groups selected from aliphatic, nitro, acetyl (i.e., -C (= 0) -CH-), or aryl groups. Each of R3 and R can independently be a hydroxy, alkoxy, halo, amino or substituted amino (such as substituted lower alkyl-amino or acetylamino or ÍÉ? 1l? 1Í ???? f ^^ - ^^^ hidroxiamino), or an aliphatic group that has 1-8 carbons and 1-20 hydrogens. When each of R3 and R4 is an aliphatic group, this may be further substituted with one or more functional groups such as a hydroxy, alkoxy, halo, amino or substituted amino groups such as those described above. The terms "aliphatic" are defined above. Alternatively, each of R3 and R4 can be hydrogen. It is well known that many 1-benzodiazepines exist as optical isomers due to chirality 10 introduced into the heterocyclic ring at the C3 position. The • Optical isomers are sometimes described as L- or D- isomers in the literature. Alternatively, the isomers are also referred to as R- and S- enantiomorphs. For simplicity, these isomers are referred to as enantiomorphs or enantiomers.
The 1-benzodiazepine compounds described herein include their enantiomeric forms as well as racemic mixtures. Thus, the use "benzodiazepine or its enantiomer," herein refers to benzodiazepine as described or represented, including all its enantiomorphs as well as 20 also its racemic mixture. From the above description, it is evident that many specific examples are represented by the generic formulas presented above. Thus, in one example, Ri is aliphatic, R2 is aliphatic, while in 25 another example, Ri is aril and R2 is a portion that participates jasas- is *.,. . ^ ^ ^ ^ ^. ^ And ^ in the hydrogen bonding formation. Alternatively, R_ may be aliphatic, and R2 may be an -NHC (= 0) -R5, or a portion that participates in hydrogen bonding, wherein R5 • is aryl, heterocyclic, -R6-NH-C (= 0) -R7 or -R6-C (= 0) -NH-R7, where R6 is an aliphatic linker of 1-6 carbons and R7 is an aliphatic , aryl, or heterocyclic. A wide variety of such combinations arising by selecting a particular group in each substituent position are possible and all such combinations are within the scope of this 10 invention. • In addition, it should be understood that the numerical ranges given through this description should be constructed as a flexible range that contemplates any possible subrange within the range. For example, the description of a group that 15 has the range of 1-10 carbons could also contemplate a group that has a subrange of, for example 1-3, 1-5, 1-8 or 2-3, 2-5, 2-8, 3-4, 3-5, 3-7, 3-9, 3-10, etc., carbons. Thus, the range 1-10 should be understood to represent the limits • outside of the range within which many possible 20 subranges are clearly contemplated. Additional examples that contemplate ranges in other contexts can be found through this description where such ranges include analogue sub-ranges. Some specific examples of the compounds of 25 benzodiazepine of this invention include: • _ ^. «L.AÍa *« .- - ^ - ^ "- .. * - -t - ^^ In summary, several benzodiazepine compounds are presented herein. Any of one or more of these benzodiazepine compounds can be used to treat a variety of dysregulatory disorders related to cell death. Such disorders include autoimmune disorders, inflammatory conditions, hyperproliferative conditions, viral infections, and atherosclerosis. In addition, the above compounds can be used to prepare 10 medications to treat dysregulatory disorders described above. The benzodiazepines described above can also be used in drug screening assays and other diagnostic methods. The broad • scope of the methodology, uses and compositions described in the present is readily apparent from a comprehensive reading of the entire description, noting that the preferred and advantageous features of some aspects are applicable to other aspects of the invention. J. Preparation of Compounds The benzodiazepine compounds described above can be prepared using either synthetic solid phase or soluble phase combinatorial methods as well as on an individual basis of well-established techniques. See, for example, Boojamra, C.G. et al. 15 (1996) J. Org. Chem. 62: 1240-1256; Bunin, B.A., et al. (1994) Proc. Na ti, Acad. Sci. USA 91: 4708-4712; Stevens, S.Y. et al., (1996) J. Am. Chem. Soc. 118: 10650-10651; Gordon, E.M. et al., (1994) J. Med. Chem. 37: 51385-1401; and North American Patents Nos. 4,110,337 and 4,076,823, which are 20 all incorporate for reference herein. For illustration, the following general methodologies are provided. 1. Preparation of 1,4-benzodiazepine-2-one compounds 25 Improved solid phase synthetic methods for the preparation of a variety of 1,4-benzodiazepma-2-one derivatives with very high total yields have been reported in the literature. See, for example, Bunin • and Ellman ((1992) J. Am. Chem. Soc. 114: 10997-10998). Using these improved methods, 1, 4-benzodiazepine-2-ones can be constructed on a solid support from these three separate components: 2-aminobenzophenones, a-amino acids and (optionally) alkylating agents, as shown in reaction scheme of Figure 1. Preferred 2-aminobenzophenones include substituted 2-aminobenzophenones, for example, the halo-, hydroxy- and halo-hydroxy-substituted 2-aminobenzophenones such as 4-halo- '-hydroxy-2-aminobenzophenones. A preferred substituted 2-aminobenzophenone is 4-chloro-4'-hydroxy-2-aminobenzophenone. Preferred α-amino acids include the naturally occurring common a-amino acids as well as α-amino acids that mimic the structures, such as homophenylalanine, homotyrosine, and thyroxine. Alkylating agents include activated and inactivated electrophiles, of which a wide variety are well known in the art. Preferred alkylating agents include the activated electrophiles of p-bromobenzyl bromide and t-butyl bromoacetate. In the first step of such synthesis, the 2-aminobenzophenone derivative, (1) of Figure 1, is attached to a support A? I.? A ^ g ^. ^ Éfttjftf > f M ^^ MjjMM ^ > i ^ aariiitA ^^ ** ^ * «« > ..--? r? -üÉiiflifrh -te "z A - * 2 £ B solid, such as a solid support of polystyrene, through either a hydroxy functional group or carboxylic acid using well known methods and employing a deblorable acid linker, such as commercially available [4- (hydroxymethyl) phenoxy] acetic acid, to produce the supported 2-aminobenzophenone, (2) See, for example, Sheppard and Williams, ((1982)) In t. J. Peptide Protein Res. 20: 451-454.) The 2-amino group of aminobenzophenone is preferably protected prior to reaction with the binding reagent, for example, by reaction with FMOC-C1 (9-fluorenylmethyl chloroformate) to produce the protected amino group 2'-NHFMOC In the second step, the protected 2-amino group is deprotected (for example, the -NHFMOC group can be deprotected by treatment with piperidine in dimethylformamide (DMF)) and the 2-aminobenzophenone does not protected is then coupled through an amide connection to a α-amino acid (the amino group of which in itself has been protected, for example, as a group -NHFMOC) to produce the intermediate (3). The standard activation methods used for general solid phase peptide synthesis can be used (such as the use of carbodiimides and active esters of hydroxybenzotriazole or pentafluorophenyl) to facilitate coupling. However, a preferred activation method employs treatment of 2-aminobenzophenone with a solution tíU¿M ^ - l. ^ MM. ~ * ^ .- ^^^^ **. i.? M ^ kmM of methylene chloride of the α-N-FMOC-amino acid fluoride in the presence of the discarded acid 4-methyl-2,6-di-tert-butylpyridine produces complete coupling through an amide linkage. This preferred coupling method has been found to be effective even for non-reactive aminobenzophenone derivatives, producing essentially complete coupling for derivatives possessing 4-chloro and 3-carboxy deactivating substituents. In the third stage, the protected amino group (the 10 which originates with the amino acid) is first unprotected • (for example, -NHFMOC can be converted to -NH2, with piperidine in DMF), and the deprotected compound was reacted with acid, for example, 5% acetic acid in DMF at 60 ° C, to produce the 1-derivative. , -benzodiazepine 15 supported, (4) • Full cyclization has been reported using this method for a variety of 2-aminobenzophenone derivatives with widely differentiated spherical and electronic properties. We In an optional fourth step, the 1,4-benzodiazepine derivative can be alkylated, by reaction with a suitable alkylating agent and a base, to produce the 1,4-benzodiazepine derivative fully supported, (5). Standard alkylation methods, for example, an excess of a strong base such as LDA (lithium diisopropylamide) or 25 NaH, can be used; however, such methods may | t |, J -. ,, *, i ^. ^ ¿^ B.iS: ra? .. ^^ result in unwanted deprotonation of other acidic functionalities and over-alkylation. Preferred bases, which can avoid over-alkylation of the # Benzodiazepine derivatives (for example, those with 5 ester and carbamate functionalities), are those which are sufficiently basic to completely deprotonate the anuided functional group, but not sufficiently basic to deprotonate amide, carbamate or ester functional groups. An example of such a base is 5-10 (phenylmethyl) -2-oxaxolidinone lithiated, which may be • reactivates with 1,4-benzodiazepine in tetrahydrofuran (THF) at -78 ° C. Following deprotonation, a suitable alkylating agent, as described above, is added. In the final stage, 1,4-benzodiazepine 15 completely derived, (6), is unfolded from the solid support. This can be achieved (along with concomitant removal of unstable acid protection groups), for example, by exposure to a suitable acid, such as a mixture of acid • trifluoroacetic, water, and dimethisulfide (85: 5: 10, by 20 volume). Alternatively, the above benzodiazepines can be prepared in the soluble phase. The synthetic methodology is illustrated by Gordon et al. (1994) J Med. Chem. 37: 1386-1401, which is incorporated herein by reference. Briefly, the methodology involves transimidating a resin 25 amino acid with 2-aminobenzophenone imines appropriately . É-rll? L-- | l-i < ? ^^ substituted to form resin bonded imines. These imines can be cyclized and attached by procedures similar to those in solid phase synthesis described above. The general purity of benzodiazepines prepared using the above methodology may be about 90% or greater. 2. Preparation of 1, -benzodiazepine-2, 5-diones A general method for the solid phase synthesis of 1,4-benzodiazepine-2, 5-diones has been reported in detail 10 by Boojamra, C.J. et al. (1996) J. Org. Chem. 62: 1240-1256. This method can be used to prepare the compounds of the present invention. This method is illustrated in Figure 2. Briefly, a Merrifield resin, for example, a (chloromethyl) polystyrene, is derivatized by alkylation with sodium 4- 15 hydroxy-2,6-dimethoxybenzaldehyde (3) to provide resin bound aldehyde ( 4) . An α-amino ester is then attached to the support derived by reductive amination using NaBH (OAc) 3 in 1% acetic acid in DMF. This • Reductive amination results in the formation of an amine 20 secondary resin-bonded (5). The secondary amine (5) was acylated with a wide variety of unprotected anthranilic acids resulting in tertiary amides attached to the support (6). This acylation can be best achieved by performing the coupling reaction in 25 the presence of a carbodiimide and the hydrochloride salt of a _M Égjiil__g | _i¡ÍÉ_? I ??,? I? - ^^^ iif p ir ifíiiÉmi? IITÍÍÍÍÍÍI Teiniiary amine. A good coupling agent is l-ethyl-8- [8- (dimethylamino) propyl] carbodiimide hydrochloride. The reaction is typically carried out in the presence of anhydrous 1-methyl-2-pyrrolidonone. The coupling procedure can be repeated once more to ensure complete acylation. The cyclization of the acyl derivative (6) can be achieved through base lactation catalyzed through the formation of an anion anion (7) which will react with an alkylhalide for the simultaneous introduction of the substituent to position 1 in the ring nitrogen heterocyclic of benzodiazepine. The lithium salt of acetanilide is a good base to catalyze the reaction. Thus, (6) can react with lithium acetanilide in DMF / THF (1: 1) for 30 hours followed by reaction with the appropriate alkylating agent that provides the bound benzodiazepine by fully derivatized support (8). Compounds (1) can be cleaved from the support in good yield and high purity using TFA / DMS / H20 (90: 5: 5). Some examples of the a-amino ester starting materials, alkylating agents, and anthranilic acid derivatives that can be used in the present invention are listed by Boojamra, supra in 1246. Additional reagents can be easily determined and either can be obtained commercially or prepared easily by an expert common in the art to reach the novel substituents described in the present invention. For example, from Figure 2, and from Boojamra, supra, one recognizes that: the alkylating agents provide the Ri substituents; the a-amino ester starting materials provide the R substituents, and the anthranilic acids provide the R4 substituent. Using these starting materials that are appropriately substituted, one arrives at the desired 1,4-benzodiazepma-2, 5-dione. The substituent R3 can be obtained by appropriately replacing the amine of the starting material a-aminoester. If steric clustering becomes a problem, the R3 substituent can be attached via conventional methods after it is isolated., 4-benzodiazepine-2, 5-dione. 3. Chirality It should be recognized that many of the benzodiazepines of the present invention can exist as optical isomers due to the quilarity wherein the stereocenter is introduced by the α-amino acid and its ester starting materials. The general procedure described above preserves the chirality of the α-amino acid or ester starting materials. In many cases such preservation of chirality is desirable. However, when the desired optical isomer of the α-amino acid or ester starting material is not available or expensive, a mixture racemic can be produced which can be separated into the corresponding optical isomers and the desired benzodiazepine enantiomer can be isolated. For example, in the case of the 2,5-dione compounds, Boojamra, supra, discloses that complete racemization can be accompanied by pre-equilibrating the hydrochloride salt of the enantiomerically pure a-amino ester starting material with 0.3 equivalents of i- Pr2EtN and the aldehyde bound to the resin for 6 hours before the addition of NaBH (0Ac) 3. He The remainder of the synthetic process described above remains the same. Similar steps may be employed, if necessary in the case of the 1,4-benzodiazepine-2-dione compounds as well. The methods to prepare benzodiazepines Individuals are well known in the art. See, for example, U.S. Patent Nos. 3,415,814; 3,384,635; and 3,261,828; which are incorporated herein by reference. By selecting the appropriately substituted starting materials in any of the In the methods described above, the benzodiazepines of this invention can be prepared relatively easily. From the above description of the invention, one skilled in the art will readily understand that the various methods of treatment, diagnosis, methods, 25 use of compounds to prepare medicines, supply of such drugs, and the making of the compounds, can be practiced in many different ways. It will also be understood that preferred aspects and features of one aspect of the invention are applicable to any other aspect of the invention. In addition, the description of this invention, coupled with one of ordinary skill in the art, can lead to additional treatments, diagnostic methods, uses, formulations and compositions. Such additional treatments, diagnostic methods, uses, formulations and compositions are within the scope of this invention, the scope of which is thus considerably broader than many examples presented herein, including those presented in the following. EXAMPLES General Methods Cell Preparation Cell lines were cultured in complete media (RPMI or DMEM containing 10% fetal bovine serum supplemented with penicillin, streptomycin, and L-glutamine) at 37 ° C, 5% C02. For activity assays, cells in log phase growth are removed and diluted to a concentration between 100,000 and 300,000 / mL. Some cells are maintained in a complete medium, while an identical aliquot is exchanged in reduced serum medium (RPMI or DMEM containing 0.2% fetal bovine serum supplemented with penicillin, streptomycin and L-glutamine) by centrifugation. Activity Assay The cells in both complete media and reduced serum media were placed in 96 well plates in 100 μL of aliquots, yielding 10,000 to 30,000 cells / well. The compound was then added to the appropriate wells in the plate (2 μL of every 50X in existence) at concentrations between 1 nM to 20 μM. The cells were then cultured overnight at 37 ° C, 5% C02). The relative cell number / cell availability was measured using standard techniques (Trypan blue exclusion / hemocytometry, MTT dye conversion assay). Example 1: Ability to Induce Cell Death Several representative individual compounds that induce apoptosis have been shown above. Of these, the most potent compounds are identified as Compound 1, which is shown in the following.
Compound 1 Compound 1 was used to induce cell death in a variety of cells using the materials and methods described above. Table 1 shows cell viability data after 18 hours of culture with Compound 1 in a reduced serum medium as described above. Table 1 • (lower numbers equal increased mortality). Nd = Not determined Example 2 MRL / MpJ-lpr / lpr mice (MRL-lpr) developed similar serological and histological manifestations of autoimmune disease as human SLE. These mice were developed by a series of cross-breeding of stresses produced up to a presented autoimmune phenotype. (Theofilopoulos A.N. and Dixon F.J. (1985) Adv. Immunol. 37: 260-390). MRL-lpr mice are characterized by the spontaneous development of systemic autoimmune disease.
This disease manifests in several physiological locations and resembles a variety of human diseases. For example, the damaged kidney in these mice is associated with high serological concentrations of anti-DNA as in human SLE. They also develop an erosive arthropathy and a lymphocytic infiltration of the salivary glands, similar to human diseases, rheumatoid arthritis (RA) and Sjórgen's disease, respectively (Theofilopoulos, supra). In general, MRL-lpr mice have a profound defect in apoptosis due to the mutation of the place of the lpr gene. (Sakata K. et al. (1998) Cl in. Immunol.Immunopa thol 87: 1-7). The defect has been linked to a mutation in the Fas receptor gene, important in apoptosis signaling in activated lymphocytes. (Watanabe-Fukunaga, R. et al. (1992) Na ture 356: 314-317). Consequently, these mice show a deep lymphoproliferation resulting in massive enlargement of the lymph nodes and the spleen. Hugely, these mice show swollen paw pads and erythematous skin lesions. Histologically, glomerulonephritis, arthritis, and inflammatory infiltration of the salivary glands are remarkable. Methods Ra tones Six-week-old female MRL-lpr mice were purchased from the Jackson Laboratories (Bal Harbor, ME). The animals were allowed to adapt to their environment for a week before the start of the treatment study. The mice were housed in a pathogen-free environment, in a specific controlled climate in a 12-hour light / dark cycle with food and water ad libitum. Once a week, weights were measured and proteinuria was examined using a colorimetric reaction (Boehringer Mannheim ChemStpp 6). 10 Treatment Regimen • Mice were randomly selected into three groups: controls receiving PBS (50 μL, qod), controls receiving DMSO (50 μL, qod), and mice receiving Compound 1 in 50 μL of DMSO (60 mg / kg qod ip for 20 mice 15 and 30 mg / kg qod ip for 10 mice). Intraperitoneal injections were given with a 30 G needle and glass syringes (Hamilton) in a dosing schedule of each additional day. The treatment started at 7 weeks of age • for control mice (those who received PBS and DMSO) 20 and at 8 or 9 weeks for the mice under treatment. At the end of the study, the blood was collected by bleeding from the tail. The mice were subsequently anesthetized by inhalation of methano and sacrificed by exsanguination by axillary dissection. The sample organs were removed 25 then by histological analysis.
J ^ J ^^. ^, ^, ... ,,,.,.,. ^^ Statistical Analysis The statistical significance analysis was made using the SPSS computer program. Unless otherwise noted, the Mann-Whitney U test (one tail) was used and the probability values > 5% were considered insignificant. Results Progression of Disease MRL-lpr mice are known to develop a kidney disease very similar to that seen in human autoimmune disease Systemic Lupus Erythematosus (SLE). The symptom of this disease is a glomerulonephritis that results in loss of kidney function and eventual death due to renal failure. A marker for the development of kidney disease is the amount of protein present in the urine. When the function of the kidney deteriorates, the glomerular filtration mechanism fails and the protein is increased. Unlike the periodicity of human disease, the murine form of lupus is progressive; Thus, once a mouse develops nephritis and the resulting proteinuria, the progress of the disease continues until death. This allows the use of protemuria measurements to monitor the progression of kidney disease in mice MRL-lpr. The development of disease in our study is ____ i__i ___ M_É _______ l followed by weekly measurement of proteinuria. Any given mouse was determined to have lupus if it had proteinuria values > 2+ (> 100 mg / dL) for 2 or more consecutive weeks. In none of the mice that met this criterion was it ever found that they had proteinuria losses below 2+. In addition, the mice that died with > 2+ of proteinuria were found to have very significant glomerulonephritis and mice that died with values < 2+ had healthy kidneys and the causes of death were not related to the autoimmune disease. Figure 3 provides analysis of disease progression for treated mice 60 mg / kg qod. Similar data were obtained with the dosing schedule of 30 mg / kg qod. As seen in Figure 3, mice treated with Compound 1 significantly delayed the attack of, and effectively treated, the disease as lupus relative to control mice (p = 0.0043). These data were supported by the observation that the BUN values for the treated animals are normal while those who received a single vehicle are with renal pains. Laboratory Diagnostics The blood of the animals was analyzed for alterations in total numbers of white blood cells (WBC) as well as the differential representation of subtypes. The mice receiving Compound 1 (60 mg / kg) had almost identical values for hematocrit, platelet count, and WBC relative to those who received a single vehicle (Table 2). Table 2. Complete Blood Count with Differential (mean ± standard deviation) # HCT, hematocrit (%); PLAT, platelets (K / μL) WBC, 10 white blood cells (K / μL) POLYS, polymorphonuclear cells (%); LYMPHS, lymphocytes (%); MONO, monocytes (%); EOS, eosinophils (%); BASO, basophiles (%). Auto antibodies The serum samples from all the mice were analyzed to determine the titre of antibodies to several autoantigens (Table 3). These antibodies are polyclonal antibodies of total serum. At the end of the study, the mice that received Compound 1 showed significantly lower titers of antibodies to ssDNA (p = 0.019) histones (p = 0.0056), and antigen La (p = 0.0265). The anti-dsDNA titers were lower in the treated mice but not statistically different from those in the • control animals (p = 0.082). There was also no difference in the 5 antibodies to the Ro antigen between the two groups of mice; however, the current absorbance measurements were very few for these ELISAs and any differences may have been masked by the sensitivity of the assay. The anti-Sm titers were observed only in a few 10 animals in both groups and no conclusion could be made regarding the differences between the groups. These anti-Sm results are consistent with the literature, which reports that only 10% of MRL-lpr mice are expected to be positive for antibodies against Sm antigen (Murphy, 15 E.D. (1981) . For data lymphoproliferation (lpr) and other single site models for murine one lupus, see Immunologic Defects in Labora tory ANIMAIS (E.M. Gershwin and B. Merchant, eds.); Vol. 2, pp. 143-173 (Plenum, New York). The differences observed in autoantibody levels are 20 found in a history of very high total IgG concentrations that do not differ statistically between the control and treatment groups (p = 0.3312). 25 *? J¿éá * t > ± e ** ~ > - ^^ M. ^^^^^ Ajb - > _ MA? AÉiM & - a < Table 3. Autoantibody titers * The titles were not available at the time of this report. Anti-histone levels are reported as OD405 values in a 1/400 dilution of serum. His tol ogía Conj un ta In addition to the lupus syndrome, the MRL-lpr mice spontaneously developed an erosive arthropathy that resembles human rheumatoid arthritis, both histologically and serologically. Arthritic lesions in these mice are characterized by inflammatory changes in the synovium and periarticular connective tissue, often accompanied by the presence of circulation of rheumatoid factors in the serum. This arthritic process is progressive and proceeds through several different stages from a mild smovitis to an erosive arthritis, which can eventually lead to a healed joint. Histologically, most 5-month-old MLR-lpr mice demonstrated synovial cell proliferation, destruction of articular cartilage and subchondral bone, infiltration of synovial stroma by inflammatory cells, periarticular inflammation (vasculitis, myositis, tendinitis, perineuritis), exudates, formation of pannus, and subcutaneous fibrinoid nodules (Hang, L. et al. (1982) J. Exp. Med. 155: 1690-1701; Koopman, W.J. and Gay, S. (1988) Scand. J. Rheumatology. Suppl. 75: 284-289). The legs of all the mice treated with the Compound 1 was examined for signs of arthritis and synovitis. Control mice (those that received a single vehicle) have a severe synovitis characterized by a marked thickening of the synovium with occasional papilla formation, hairy configurations. Typically, the smovial pathology is a result of the proliferation and synovial cellular infiltration of the synovial stroma by inflammatory cells. In a substantial percentage of the control mice, the disease process is accompanied by pannus formation and erosion of the articular surface (both the articular cartilage and the subchondral bone). In contrast, the mice under treatment were found to have a milder synovitis as well as few erosions and limited pannus formation (Table 4). The character of the disease in animals receiving Compound 1 was much less aggressive with less synovial cell proliferation and inflammatory infiltration. Of greater interest, it was observed that the mice under treatment had a decreased degree of periarticular inflammation. The combination of these results suggests that Compound 1 slows the disease process t .... ¡. í ^ _á_i -_... ** * .. *, *. , ....,. ^. ^^ aM ^ A ^ - .. ^^^^ arthritic that typically destroys the joints of the MPK-Ipr mice. Table 4. Synovial Pathology • P value determined by cross-tabulation and 5 chi-square analysis Hypersensitivity of the Delayed Type (DTH) Mice treated with Compound 1 (60 mg / kg) showed no difference in the DTH response to TNBS compared to control mice ( Figure 4C). Importantly, neither of the two groups of animals showed a significant swelling of the paw pads. • following the antigen challenge. This phenomenon has been documented and old MRL-lpr mice (> 10 weeks) are expected to have a decreased in vivo T cell response to stimuli as evidenced by the absence of a DTH response. See Okuyarna, H. et al. (1986) Cl in. Exp. Immunol. 63: 87-94, and Scott, C.F. et al. (1984) J. ^^ -_- Uto _.-_- »^ a. i ».- **. ' > ^^.-.- ^^ J-il ^^^^ J ^. ^ - a - ^ Immunol. 132: 633-639. However, suppression of T cells may result in a rescue of the DTH response. See, Okuyama H. et al. (1989) Int. Arch. All ergy appl. Immunol. 1588: 394-401. Such a rescue was not observed in this treatment protocol of the study. These data suggest that Compound 1 does not alter the function of T cells. Figures 4A-4C show the results of the Swelling experiment of the Paw Bushings. Immune Cell Function Thymidine incorporation assays to use stimulated and unstimulated T and B cells led to the determination of whether Compound 1 affects in vitro lymphoproliferation. At approximately a concentration of 10 μM, no effect on lymphocyte proliferation was observed. Example 3. Compound 1 as a lympho-toxic agent Methods Animals and experimental design: NZB / W female mice (Jackson Labs) were housed in environmentally controlled, pathogen-free rooms operated by the University of Michigan's Unit for Laboratory Animal Medicine with 12 hours of light-dark cycles and were given food and water ad libitum. Randomly distributed mice in the treatment and control groups. All mice were dosed through intraperitoneal injections using a Hft.-..--. M ffi ^ _ ^^ AJ ^ .- »- -_MMl ^^ Hamilton repeat with glass microliter syringes and 30 gauge needles. Control mice received the vehicle (50 μL of aqueous DMSO) and the treatment mice • received Compound 1 dissolved in vehicle. The weight of 5 animals was determined weekly, and the dosing schedules were adjusted again later. Blood Collection / Temporary: Peripheral blood was obtained from the tail vein of all mice for analysis of complete blood counts and collection 10 of serum. The blood was first allowed to clot # room temperature for 1 hour, and then overnight at 4 ° C. The serum was separated from the clot formed by centrifugation (6 minutes, 16,000 x g). A section of the spleen was removed aseptically for the preparation of suspensions 15 of a single cell. Samples of the following organs were preserved in 10% buffered formalin; heart, liver, lung, spleen, kidney, small intestine, reproductive system, salivary glands, thymus, mesenteric and axillary lymphoid nodes, and skin. Additional sections of the kidney and spleen 20 were preserved by instantaneous freezing in OCT. The bone marrow spots were prepared from each femur. His tology: All histological determinations were done in a blind way by a pathologist. The sections fixed with formalin were cut and stained with Hematoxylin and eosin (H &E) using standard protocols (Luna, L.G., in: Manual of Histological Staining Methods of the Armed Forces Institute of Pathology, McGraw-Hill, New York (1960)). Deposition of the immune complex in the kidneys was evaluated by direct immunofluorescence using frozen sections stained with FITC-conjugated goat anti mouse goat (Southern Biotechnology Associates, Birmingham, AL) and C3 (Cappel-organon Teknika, Durham, NC). The degree of lymphoid hyperplasia was classified on the 0-4 + scale. TUNNEL DYEING: Frozen spleen sections (4 pm thick) were tested by DNA strand breaking using the In Situ Cell Death Detection (Roche Molecular Biochemicals) equipment according to the manufacturers' protocols. Sections were analyzed using a 0-4 + scale. The sections were evaluated blindly and a classification was assigned on the basis of the amount of positive TUNNEL dyeing. Fluorescence analysis of lymphocyte populations: Single-cell suspensions were prepared by a tear that separates the spleen in the medium, followed by removal of red blood cells with isotonic lysis buffer (Kruisbeek, AM, en: Current Protocols in Immunology, eds., Coligan, JE et al., Pp. 3.1.2-3.1.5, John Wiley &Sons, Inc. (1997)). 106 cells were stained at 4 ° C with anti-Thy 1.2 fluorescently conjugated (Pharmingen, clone: 53-2.1, 1 μg / mL) and / or anti-B220 (Pharmingen, clone: * ^,. * .. ...,., .., i ^^ - «_ ^ a ^« - ^^, RA3-6B2 1, μg / mL) for 15 minutes. In the samples stained for detecting outer membrane phosphatidylserine, the cells were then incubated with Annexin V and Pl conjugated FITC according to the manufacturer's protocols (Roche Molecular B Lochemicals). The cells were analyzed in a Coulter ELITE flow cytometer. For each sample, at least 10,000 cases were counted. Anti-DNA Serum: Titers were determined by direct ELISA as previously described (Swanson, P.C. et al., J. Cl in. Invest. 97: 1748-1760 (1996)). The detection of anti-DNA IgG used an alkaline phosphatase conjugated goat anti-mouse IgG (only H chain) secondary antibody (1/1000 dilutions, SIGMA). To convert the absorbance readings into the titers, the pooled serum of the eight-month-old unmanaged NZB / W females was used as a standard reference which arbitrarily assigned a value of 1000 U / mL. Serum immunoglobulin: Concentrations were determined by ELISA capture. The goat anti-mouse Ig (Southern Biotechnology Associates) was diluted to 10 pg / mL in PBS and coated overnight at 4 ° C on Immulon II microtiter plates. Otherwise, ELISAs were performed as previously described. To convert absorbances at the concentrations, a standard carva was generated using an immunoglobulin reference serum i ^^ g of previously quantified mouse (ICN Biomedicals, Aurora, OH). Neither blood urea triogen (BUN) and complete blood count (CBC): BUN serum measurements were conducted by the pathology laboratory of the University of Michigan Hospital clinic. The CBC analyzes were conducted by laboratory diagnosis of the University of Michigan's Unit for Laboratory Animal Medicine. The automated counts determined by a Hemavet 15 OR were confirmed by visual examination of blood spots. Serum levels of 1, 4 -benzodia zepine: Serum samples from mice injected with Compound 1 were precipitated with acetone (5X volume, -20 ° C, 10 minutes). After centrifugation (16000 xg, 10 minutes), the supernatant containing Compound 1 was concentrated in vacuo, and then extracted from any remaining protein using a Sep-pak C18 column (Waters Corp.) running a gradient of 10O stage of acetonitrile in water at 100% acetonitrile. Eluting material in the organic fraction was concentrated in vacuo, and then analyzed by reverse phase HPLC using a Phenomenex C18 column. Peak areas were determined using a Shimadzu mtegrador and referenced to a standard curve. Statistical Analysis: Statistical analyzes were conducted using the SPSS software package. The ÍÉr_ t IT- heh -? *** '* - *' "• * • '. ___ ____ __ ^ M_iMdÍÍte tests Mann-Whitney U and chi-block were used for histological and clinical data Student t test was used to citométpcos flow data. correlations were assessed by ANOVA. Example 4 a series of experiments were conducted to characterize the nature of cell death caused by Compound 1 in terms that could point toward the death mechanism. Compound 1 treated cells stained positive in the TUNNEL assays that suggest the result of the treatment in free ends of DNA (Gorczyca, W. et al., Leukemia 7: 659: 670 (1993)), an aspect of apoptosis. The activity of Caspase and mitochondrial permeability are two central components of the intracellular killing machinery (See, Zamzami, N, et al., J. Exp. Med. 183: 1533-1544 (1996) and Los, M. et al. ., Immuni ty 10: 629-639 (1999)). Cyclosporin A (CsA) regulates the mitochondrial permeability transition (MPT) (Zoratti, M. and Szabo, I., Biochim Biophys Acta 1241:.. 139-176 (1995)), the protective cell death paths they depend on the release of apoptogenic factors of the mitochondria. CsA provides dramatic protection against Compound 1, such that 90% of the cells that could have been eliminated by Compound 1 survive when CsA (10 μM) is added to the culture (Figure 6B). The pre-treated cells * With z-VAD-fmk (20 pM), an inhibitor of non-specific caspase (Garcia-Calvo, M. et al, J. Biol Chem 273:... 32608-32613 (1998)), protects cells to a lesser grade: viability in pre-treated z-VAD-fmk cells is 25% greater than cells treated with Compound 1 alone. Together, these data suggest that Compound 1 that induces death of ZNB / W lymphocytes requires MPT while caspase activation is of secondary importance. To determine if the effects on lymphocytes in vitro also occur in vivo, female NZB / W mice of 8 months of age were dosed with Compound 1 (60 mg / kg PI daily). This dose resulted in peak serum titers of 10 μM (1 h after injection) but with levels of 1-2 μM (18-24 hours after injection). After 7 days, the mice were sacrificed and their spleen cells were examined by flow cytometry. The splenocyte viability, measured in the Pl exclusion base, was significantly decreased in injected animals treated against control as shown in Table 5, below. Table 5 Sodnocyte flow cytometric measurements after in vivo treatment. • In this experiment, eight-month-old NZB / W mice were divided into treatment and control groups of 5 seven mice each. The mice in treatment were dosed with Compound 1 (60 m kg qd) and the controls received vehicle (50 μL DMSO qd) for 7 days. The total viability of the splenocytes isolated from each mouse was determined in the exclusion basis Pl. The decrease in • 10 viability is less than that observed after in vitro treatment because intervention processes (i.e., cell recruitment, proliferation and cleaning of early apoptotic cells by the reticuloendothelial system (RES)) decrease death splenocyte 15 detectable. Apoptotic cells are shown as a percentage of B and T cells that exclude Pl. Mixed splenocytes were isolated from the treated animals and then placed in cultures briefly (2 x 10 6 cells / mL for 3 hours in RPMI containing 10% FBS) to allow 20 expression of the apoptotic phenotype in the absence of cleaning RES. Subsequently, the cells were stained with Annexin V-FITC, Pl and monoclonal antibodies specific for either Thy-1.2 or B 20. Each value is based on the flow cytometric analysis of 20,000 viable cells.
Tra MENT ra tones NZB / W with Compound 1 A therapeutic dose, current treatments for lupus organ threatened severely depleted all lymphoid cells, resulting in serious side effects associated 5 with immunosuppression (Donadio, JV and Glassock, RJ, Am. J. Kidney Dis. 21: 239-250 (1993)). The B cell selectivity coupled with the moderate total effect on splenic lymphocyte viability is distinguished from Compound 1 from these treatments. Therefore, Compound 1 represents 10 a compound directed at a new class of agents • lymphotoxics possibly with a favorable beneficial toxicity profile. To determine the effect of Compound 1 on autoimmune nephritis and how it effects the long-term dosing of the immune system, a treatment study 15 longitudinal was conducted using female NZB / W mice. In this study, the mice received intraperitoneal injections of Compound 1 (n = 25, 60 mg / kg) or vehicle (n = 20) every day for 3 months from 6.5 to 9.5 months of age. f This time lapse represents a point after the attack 20 of glomerulonephritis and continues through 50% of the mortality point (Theofilopoulos A.N. and Dixon, F.J., Adv. Immunol., 37: 269-390 (1985)). The mice were sacrificed periodically and examined for the impact of Compound 1 on nephritis and the peripheral immune system. 25 Microscopic examination of kidney tissue __i ^ * -, - J .- »- --..-" .- a- ^ > , ...- ^^., > _.- ^? ^ ... ^ a .., .. ^^ revealed that the treatment with Compound 1 is protective through the course of treatment. Kidney sections taken from the control mice indicated a diffuse, proliferative glomerulonephritis with proliferation of all cellular elements and occasional curly wire formation consistent with a histopathological average rating of 3+ (Figures 6 and 7). In contrast, the treatment mice have many smooth changes with less cell proliferation and curled wire formation and a score of 1+ (p <0.002). As seen in Figure 6, administration of Compound 1 reduces glomerular IgG and C3 deposition throughout the course of treatment (IgG, p <0. 003; C3, p <0.04). The degree of deposition with an individual mouse correlates strongly with histopathological classification: mice with reduced Ig deposition showed less glomerular damage (p <0.002). Mechanically, this finding is consistent with lymphoid cell death in vivo, presumably leading to few (pathogenic) B cells. The histological differences observed between the treatment and the control groups were confirmed by clinical measurements of renal function. At the time of sacrifice, 85% of the control mice had abnormally high blood nitrogen urea (BUN) levels (2 30 mg / dL) (Gordon, C. et al., Clin.Immuno 1. Immunopa.T. 52: 421 - 434 (1989)), while 31% of the mice under treatment have high BUN (p <0.007). Similarly, 39% of control mice have significant proteinuria (> 100 mg / dL) (Adelman, NE et al., J. Exp. Med. 158: 1350-1355 (1983)) while only 18% of the treated mice developed this finding (p <0.11). For each mouse, the histological classification is strongly correlated with proteinuria and BUN levels (p <0.002); mice with less severe disease have lower proteinuria and BUN titers. These data confirm that histological measurements of nephritis reflect the functional status of the kidneys. Together, these findings indicate that Compound 1 protects NZB / W mice from the progression and effects of autoimmune glomerulonephritis. Compound 1 reduces the number of cells B NZB / W mice develop lymphoid hyperplasia related to the disease marked by an expansion of B cells that coincides with the increase in serum titers autoantibody (See, Theofilopoulos, AN and Dixon, FJ, Adv. Immunol., 37: 269-390 (1985) and Ermak, TH et al., Lab. Invest. 61: 447-456 (1989)). The spleens of the control mice reflected this phenomenon: an expansion of the white pulp was observed causing distortion of the architecture of the normal spleen and a reduction in the red pulp (Figure 10). The mice that received Compound 1 have a more balanced normal red and white pulp. Sliced multiple sections of all the treatment and control animals revealed a statistically significant reduction of lymphoid hyperplasia in the treatment group (2+ versus 3+ on a scale of 0-4 +; p < 0.02). The constitution of the splenic lymphoid cell compartment after 6 and 12 weeks of treatment was analyzed by flow cytometry. As shown in Table 6, Compound 1 reduced the B cell fraction by 10-15% relative to the control mice (p <0.05), while the fraction of the T cells remains unaffected by the treatment.
Table 6 Cytometric flow determination of splenic lymphocyte populations occurs after longitudinal treatment This difference can be explained either by an effect on lymphocyte proliferation or by reduction | i-MiAi -__ ÍMk? éMz ____ «-....,. «> ^ -ai a-. «_» - ~ * u ~.,. *. A ^^ MÍ ^^ JM > JtMM¿ > JM ^ ^ J ^^ JMi ^ _ ** _. X .....
Splenocy proliferation in vi tro, the last explanation is favored. No significant increase in the fraction of T cells was observed, suggesting the elimination of T cells that occurs to a lesser extent than the elimination of 5 B cells, which is consistent with the findings of in vitro studies and short dosing experiments. term previously presented. To directly determine the effect of Compound 1 on lymphoid cell death in vivo, the tissue 10 splenic was analyzed for evidence of cell death • using the TUNNEL reaction. In the sections of 10 representative control mice, the number of positive cells TUNNEL to less than 1% of the total number of cells and staining were randomly dispersed throughout the entire 15 section. In contrast, mice treated with Compound 1 possess up to 5-fold increase in the numbers of positive TUNNEL cells (approximately 3+ staining) in 50% of the sections (p <0.05). Unlike the sections of the control mice, the positive TUNNEL cells in these 20 sections showed prominent clustering (Figure 10). The combined TUNNEL assay and immunohistochemical staining for the B cell surface marker (B220) indicated that the cells demonstrating drug-induced TUNNEL positively are B cells (Figure 10). 25 The action of Compound 1 against B cells and the improvement in kidney disease mediated auto-antibody predicts a decrease in autoantibody titers. The serum obtained during the course of the treatment was tested for total IgG as well as the anti-dsDNA. The total immunoglobulin levels are not altered during the course of treatment (eg, after 9 weeks of treatment, control = 4.0 ± 2.2 mg / mL, Compound 1 = 4.0 + 1.3 mg / mL, p> 0.4). In contrast, the anti-dsDNA titers were reduced after 3 weeks of treatment with Compound 1 (control = 733 + 546 U / mL; Compound 1 = 496 + 513 U / mL, p <0.06). However, for about 9 weeks the anti-dsDNA treatment levels in treated mice are indistinguishable from the contoles (control = 812 + 695 U / mL; Compound 1 = 944 + 546 U / mL, p> 0.3). Early fall in anti-dsDNA in the context of total IgG without change coupled with the observation that glomerular Ig deposition is reduced throughout the entire course of treatment, suggests that pathogenic cells may have an improved sensitivity to Compound 1. Given the progressive nature of the disease in NZB / W mice, it is likely that the eventual elevation of anti-dsDNA corresponds to a point characterized by increased B cell proliferation, for example, as can be seen in Table 6. This hypothesis is also supported by the observation that dosed twice the dose of the compound which results in a sustained reduction of anti-dsDNA with the improved disease approximately equivalent to that seen with the lowest dosage. In summary, these observations link treatment with Compound 1 to ameliorate the disease, as reflected by renal histology, renal function, autoantibody, and C3 deposition, and provide evidence that this therapeutic effect matches the ability of the compound to selectively induce cell death in B cells. Compound 1 is non-toxic Animal tissues were examined for evidence of toxicities commonly associated with cytotoxic drugs. Compound 1 does not affect the gross weight of the animal and mice receiving Compound 1 do not have altered grooming practices or markedly different behaviors. Microscopic examination of heart, liver, lung, salivary glands, small intestine, and uterus showed no significant architectural differences between the control and treatment groups. In addition, comparing the control and treatment groups revealed that Compound 1 does not increase the number of apoptotic cells in non-lymphoid organs. No evidence of increased pneumonitis was observed in the lungs of the treated animals, which is an important indicator of common infection. ««. ",.» ^ - t --- < -A _J-i_fc-_J --_-. ^^^? ^^^^^ j ^? ák? ^ m? ^? observed in animal studies using cytotoxic drugs (Horowitz, R.E. et al., Lab. Invest. 21: 199-206 (1969) and Hahn, B.H. et al., Arthri tis Rheum. 18: 145-152 (1975)). The loss of excess pneumonitis in treated animals is strong evidence against a significant immunosuppressive effect. The examination of spots in the bone marrow does not reveal differences between the treatment and control animals with respect to total cellularity or in the proportional representation of specific myeloid and 10 erythroid precursors. Peripheral blood counts and • analysis of differential white blood cells did not reveal a decrease in platelets, granulocytes, lymphocytes, or hematocrit. In the aggregate, no evidence of generalized immune suppression or other toxicities are detected in 15 mice treated with Compound 1. Example 5 For model neuroblastoma in mice, mice were transfected with the SKNAS human neuroblastoma cell line, to cause the cells to overexpress the 20 neuroblastoma associated with the human oncogene N-myc. The resulting cell line is designated as D2. These cells form tumors when they are xenoinjertan in T cell deficient athymic mice; thus, a relevant animal model of human neuroblastoma is provided. 25 In vitro tests of D2 cells are ______ if ____-_____? ___ É -_? ri? i_l__tfi__i were conducted to determine their sensitivity to benzodiazepine. D2 cells were coated in 96-well tissue culture plates at a density of 10,000 cells per well in a culture medium (DMEM, 10% V: V of 5 heat-inactivated fetal bovine serum (FBS), 100μ / ml penicillin, 100 μg / ml streptomycin, 290 μg / ml glutamine) and cultured (37 ° C, 5% C02) overnight. Subsequently, the culture media was exchanged with media containing 1% FBS. Solvent Control (dimethylsulfoxide (DMSO), final concentration 1% V / V) or benzodiazepine at concentrations of 2.5-20 μM were added. After 18 hours, cell viability was measured using the MTT assay as previously described in this application. Figure 11 showed that the 15 benzodiazepine eliminates D2 cells in a dose response form. To test the effect of benzodiazepine on neuroblastoma tumor growth, 1x10 D2 cells were aseptically inoculated into the thigh musculature of each 20 one of eight female six-week-old nu / nu mice (Jackson Labs). Starting one week after the tumor cell inoculation, 4 mice were dosed with DMSO (20 μl injected into the peritoneal cavity each day) and 4 mice were dosed with benzodiazepine (2.5 mg dissolved in 25 20 μl of DMSO injected into the peritoneal cavity every day).
The mice were evaluated regularly to develop tumor and once present the size of the main tumor was measured every day. Table 7 shows that in mice that were formed • tumors, treatment with benzodiazepine significantly slowed the growth of the tumor. Table 7 # The administration of slow Benzodiazepine velocity of neuroblastoma tumor growth in nu / nu mice (p <0.02). Specifically, tumors in control mice are • increased in volume 5 times over an average of a period of 4 days, while 12 days were required for the same increase in tumor size in animals treated with benzodiazepine (p <0.02). These findings support the 15 claim that benzodiazepine is capable of treating human malignant disease in a mouse model. Besides, the Benzodiazepine has specific activity against human neuroblastoma in vitro and in vivo. Example 6 In other lines of experiments, it was sought to determine whether benzodiazepine is able to eliminate tumor cells that are otherwise resistant to the standard chemotherapy drugs present. Ovarian cancer provides an excellent model to study the problem of chemoresistance where the treatment fails, they are 10 commonly attributed to the emergence of cells • resistant to chemotherapy. The human ovarian cancer cell line A2780 is known to contain p53 of the wild type; expressing low levels of survival factors bcl-2 and bcl-xL; and is sensitive to treatment 15 with cis-platinum (II) diamine dichloride (CDDP), a standard chemotherapy for ovarian cancer treatment. These cells were transfected with an expression vector that encodes human bcl-xL, a survival factor that when overexpressed is linked to the development of resistance to 20 chemotherapy. These transfected cells are designated 2B1, and the transfected controls of the empty vector are designated vector only. A third ovarian cancer cell line, designated SK0V3, was also obtained. This cell line is characterized as: 1. Deficient in the expression 25 p53 wild-type; 2. Express high levels of bcl-x_ endogenous and 3. Relatively resistant to the cytotoxic actions of CDDP. Each of these cell lines was maintained using standard tissue conditions in complete media composed of RPMI, 10% FBS, 100 U / ml penicillin, 100 μg / ml streptomycin, 290 μg / ml glutamine. Each type of cell was coated in a series of separate wells in 24-well tissue culture plates at 50,000 cells per well. Approximately 24 hours after the coating, the media was exchanged to contain the same culture media made with only 2% FBS. At this point any control solvent (DMSO, 1% V / V), the increased concentrations of Compound 1 (4-20 μM), or the increased concentrations of CDDP (6.7-66.7 μM) were added to the cells. After twenty-four hours of culture all the cells present in each well were removed using trypsin-EDTA and mixed with propidium iodide (final concentration 1 μ / ml). After incubating the cells 20 minutes were analyzed by flow cytometry (Coulter FACS Calibur) to determine cell death at the bases of the integrity of the plasma membrane measured as the fraction of cells that had received propidium iodide. Figure 12 demonstrates that the predicted pattern of chemosensitivity and resistance to CDDP (A2780 and sensitive vector; 2B1 and resistant SKOV3) was observed. Figure 13 ^^ j ^^ jg | ft¡ ÉiÉ __________________ ttÍ_ | Í ^^ fe @ ÍtíM demonstrates that benzodiazepine eliminates each of these types, regardless of the CDDP resistance. In addition, benzodiazepine removes ovarian cancer cells that are resistant to standard chemotherapy. - In addition, benzodiazepine removes tumor cells that express high levels of survival factors (bcl-xL), as well as those that are deficient in p53 expression. Although the preceding invention has been described in some detail by way of illustration and example for 10 purposes of clarity of understanding, will be evident by • Those skilled in the art that certain changes and modifications will be practiced. Therefore, the description and examples should not be construed as limiting the scope of the invention, which is delineated by the 15 attached claims.
F

Claims (1)

  1. CLAIMS 1. A method for treating a condition associated with dysregulation of the cell death process in a subject, characterized in that it comprises administering to the subject an effective amount of a benzodiazepine compound. The method according to claim 1, characterized in that the benzodiazepine does not bind to a central benzodiazepine receptor and binds only with low affinity to a peripheral benzodiazepine receptor. 3. The method according to claim 1 or 2, characterized in that benzodiazepine induces apoptosis in a low serum assay. 4. The method according to claim 1, characterized in that the condition is not a chronic inflammatory condition. 5. The method according to claim 1, characterized in that the benzodiazepine is a compound having the structure: ., ^,., _ «_...... * -tff. , 1GÍÍÉÍML ______ L ________ L ____ Í _______ ^^ or its enantiomer, where Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2 -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -Rβ-NH-C (= 0) -R7 or -R6-C (= 0) -NH-R-;, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, 10 hydroxy, alkoxy, halo, amino, lower alkyl-amino • substituted, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 6. The method according to claim 1, characterized in that the benzodiazepine is a compound having the structure: & i ^^ ____________ g__É ______ i ________- ii-ii_a or its enantiomer, where Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) - R7 or -Re-C (= 0) -NH-R7, wherein Re is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, 10 hydroxy, akoxy, halo, amino, lower alkyl-amino • substituted, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aplo, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 7. The method according to claim 1, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein, 20 Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -Re ~ NH-C (= 0) - R or -R6-C (= 0) -NH-R7, wherein R ^ is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amine, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 8. The method according to claim 1, characterized in that cell death is apoptotic. 9. The method according to claim 1, characterized in that the cell death is necrotic. 10. The method according to claim 1, characterized in that the dysregulation of the cell death process is caused by the interruption of the FAS path. 11. The method according to the claim I, characterized in that the condition is an autoimmune disease. 12. The method in accordance with the claim II, characterized in that the autoimmune disease is a disease selected from the group consisting of lupus ^^^ systemic erythematosus, rheumatoid arthritis, Sjogren's syndrome, graft-versus-host disease and myasthenia gravis. 13. The method according to claim 1, characterized in that the condition is a chronic inflammatory condition. The method according to claim 11, characterized in that the chronic inflammatory condition is psoriasis, asthma, or Crohn's disease. 15. The method of compliance with the claim 1, characterized in that the condition is a hyperproliferative disorder. 16. The method according to claim 15, characterized in that the hyperproliferative disorder is a neoplastic condition. 17. The method according to claim 15, characterized in that the hyperproliferative disorder is selected from the group consisting of B-cell lymphoma, T-cell lymphoma, cancer, chemoresistance cancers, disorders related to deficient p53 expression, and related disorders. overexpression of endogenous bcl-xL. 18. The method according to claim 1, characterized in that the condition is induced by a viral infection. 19. The method of compliance with the claim .A * -M.i, «M ^! -. ... *? ^ .., *,? ^, .. ^ .. £ - 16, characterized in that the viral infection is caused by a virus selected from the group consisting of herpes virus, papilloma virus and Immunodeficiency Virus Human (HIV)). 20. The method of compliance with the claim 1, characterized in that the condition is atherosclerosis or osteoarthritis. 21. The method according to claim 1, characterized in that it further comprises co-administering one or more additional agents to the subject. 22. The method according to claim 21, characterized in that the additional agent is a chemotherapeutic agent or radiation. 23. The method according to claim 1, characterized in that the compound is administered orally, parenterally, topically or intranasally. 24. A method for treating an autoimmune disease in a subject characterized in that it comprises administering to the subject an effective amount of a benzodiazepine compound. 25. The method according to claim 24, characterized in that the benzodiazepine does not bind to a central benzodiazepine receptor and binds only with low affinity to a peripheral benzodiazepine receptor. 26. The method of compliance with iíb.i.? ?? ^? í? ^ LA? im.i .... * -. ^ a ^, ..... ^., .. _-.._ ^ «- L-yi ^ claims 24 or 25, characterized in that benzodiazepine induces apoptosis in a low serum assay. 27. The method according to claim 24, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R &-NH-C (= 0 ) -R7 or -R? -C (= 0) -NH-R7, wherein Re is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. The method according to claim 24, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -Re-NH-C (= 0) - R 15 or -R 6 -C (= 0) -NH-R 7, wherein R 6 is an aliphatic linker of 1"6 carbons and R 7 is aliphatic, aryl, or heterocyclic, and each of R 3 and R is independently hydrogen, hydroxy , alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group which } ^ ^ * »^ - * ^». ** ~. *. . ^^^^ * ^ - .. ^^, ^ .. ^^ .. a ^^ t ^ W? FlTr? R? Ll1 Étím has 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 29. The method according to claim 24, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) -R7 or -R6 ~ C (= 0) -NH-R7, where Re is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and • each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. WptÉiiftiftif * ^^ - "" ^ 30. The method according to claim 24, characterized in that the autoimmune disease is a disease selected from the group consisting of lupus. • systemic erythematosus, rheumatoid arthritis, 5 Sjogren, graft-versus-host disease and myasthenia gravis. 31. The method according to claim 24, characterized in that it further comprises co-administering one or more additional agents to the subject. 10 32. The method of compliance with the claim • 31, characterized in that the additional agent is an immunosuppressant. 33. A method for treating a chronic inflammatory condition in a subject characterized by 15 comprises administering to the subject an effective amount of a benzodiazepine compound. 34. The method according to claim 33, characterized in that the benzodiazepine does not bind to a • central benzodiazepine receptor and binds only with 20 low affinity to a peripheral benzodiazepine receptor. 35. The method according to claim 33 or 34, characterized in that benzodiazepine induces apoptosis in a low serum assay. 36. The method according to claim 25 33, characterized in that the benzodiazepine is a compound which has the structure wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -Re-NH-C (= 0) - R7 or R6-C (= 0) -NH-R7, where R? is an aliphatic linker of 1-6 10 carbons and R7 is aliphatic, aryl or heterocyclic; and each of R3 and R is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; 15 or a pharmaceutically acceptable salt, prodrug or derived from it. 37. The method according to claim 33, characterized in that the benzodlazepine is a compound having the structure: or its enantiomer, wherein, R is aliphatic or aplo; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -Rs, or a portion that participates in the formation of hydrogen bonding, wherein Rs is aryl, heterocyclic, -R6-NH-C (= 0) - R or -R6-C (= 0) -NH-R7, where R? is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. .-----. and -, ^. ^^ -. * • - & ** »* * .------- ° ^^ - é ---- É-l - É -! -! - í - $ - ffi 38. The method according to claim 33, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein Ri is aliphatic or aryl; • R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -Rg-NH-C (= 0) -R7 10 or -Rg-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group which • 15 has 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 39. The method according to claim 33, characterized in that the chronic inflammatory condition is psoriasis, asthma, or Crohn's disease. In addition, the method according to claim 33, characterized in that it further comprises co-administering one or more additional agents to the subject. • 41. The method according to claim 5, characterized in that the additional agent is an anti-inflammatory agent. 42. A method for treating hyperproliferative disorder in a subject characterized in that it comprises administering to the subject an effective amount of a 10 benzodiazepine compound. • 43. The method according to claim 42, characterized in that the benzodiazepine does not bind to a central benzodiazepine receptor and binds only with low affinity to a peripheral benzodiazepine receptor. 44. The method according to claim 42 or 43, characterized in that benzodiazepine induces apoptosis in a low serum assay. 45. The method according to the claim Ww 42, characterized in that benzodiazepine is a compound 20 that has the structure: Í «-_. & -. Af _« - «¿-l - ^ .-- fc- ^ •% • or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) - R or Rg-C (= 0) -NH-R7, wherein Re is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl or heterocyclic; and each of R3 and R4 is independently hydrogen, Hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 46. The method according to claim 42, characterized in that the benzodiazepine is a compound tütlini-w iin- iHirtf? Ílt "- - '*' ff f Tf ** 1fH that has the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; • R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0 ) -R or -Rg-C (= 0) -NH-R7, wherein Rg is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each R3 and R is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group that • has 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 47. The method according to claim 42, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -Rs, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) -R • or -Rg-C (= 0) -NH-R, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. • 15 48. The method according to claim 42, characterized in that the hyperproliferative disorder is a neoplastic condition. 49. The method according to claim 42, characterized in that the hyperproliferative disorder is 20 selects from the group consisting of B-cell lymphoma, ? - ^ -A - * - »^» - ^^ - ^ tttMU-Ji --- ^^^. ^ .. ^. I, ^^^! ^^^ T cell lymphoma, cancer, cancers of chemoresistance, disorders related to poor p53 expression, and disorders related to the overexpression of endogenous bcl -? _. 50. The method according to claim 42, characterized in that it further comprises co-administering one or more additional agents to the subject. 51. The method according to claim 50, characterized in that at least one additional agent is 10 a chemotherapeutic agent, or radiation. F 52. A method for treating a condition associated with dysregulation of the cell death process in a subject, characterized in that it comprises administering to the subject an effective amount of a benzodiazepine compound, in 15 where the condition is induced by a viral infection. 53. The method according to claim 52, characterized in that the benzodiazepine does not bind to a central benzodiazepine receptor and binds only with F low affinity to a peripheral benzodiazepine receptor. 54. The method according to claim 52 or 53, characterized in that benzodiazepine induces apoptosis in a low serum assay. 55. The method according to claim 52, characterized in that the benzodiazepine is a compound 25 that has the structure: i ^. ^, ^^^^ ._ i. ff). ^ rn r »^ ttM ^^ or its enantiomer, where, • Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -Rs, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -Rg-NH ^ C (= 0) - R7 or R6-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl or heterocyclic; and each of R3 and R4 is independently hydrogen, 10 hydroxy, alkoxy, halo, amino, lower alkyl-amino # substituted, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 15 56. The method according to claim 52, characterized in that the benzodiazepine is a compound ftr "" - -t (Mtm? M? tß ^ MáíáÉii imMu ^ »Mu? l ^^ Í ^^^^ that has the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; • R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0 ) -R7 or -R6-C (= 0) -NH-R7, wherein R & is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group that • has 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 57. The method according to claim 52, characterized in that the benzodiazepine is a compound having the structure: "T 'j ^ f"' ^ "--- • 11 ^^ ^^^^. ^. ^^». ^ - ^ - a - ^ «^ - ^^. - ~ ^ > < - **** t «¿* ~ ..J3t? .. U. or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) -R or -R6-C (= 0) -NH-R7, wherein RQ is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, lower substituted-amino alkyl, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. • 15 58. The method according to claim 52, characterized in that the viral infection is caused by a virus selected from the group consisting of herpes virus, papilloma virus and Human Immunodeficiency Virus. 20 59. The method of compliance with the claim 52, characterized in that it further comprises co-administering one or more additional agents to the subject. 60. The method according to claim 59, characterized in that the additional agent is an antiviral agent. 61. A method for promoting cell death characterized in that it comprises contacting a cell or tissue with an effective amount of a benzodiazepine compound. 62. The method according to the claim 61, characterized in that the benzodiazepine does not bind to a central benzodiazepine receptor and binds only with low affinity to a peripheral benzodiazepine receptor. 63. The method according to claim 61 or 62, characterized in that benzodiazepine induces apoptosis in a low serum assay. 64. The method according to claim 61, characterized in that the benzodiazepine is a compound having the structure: • '"^ T'irrT-fr - ^ - ff | fi - ^ -» "-» ^ «» ^ r - ^ - "" - • a? Ff ^ ff- * or its enantiomer, where, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) - R7 or -Rd-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 65. The method according to claim 61, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -Rg-NH-C (= 0) - R7 or -Re-C (= 0) -NH-R7, wherein Re is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 66. The method according to claim 61, characterized in that the benzodiazepine is a compound having the structure: f "**" * "- | ff f * ff '" "• ftt-'t ^ ^ --- ^^^ - ^^^ - ^ *«' ^ '* ^^ - ^ - & -l * ~ XX? To its enantiomer, where Ri is aliphatic or aryl, R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bond, wherein R5 is aryl, heterocyclic, -RS-NH-C (= 0) -R or -R6-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic, and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic, or a pharmaceutically acceptable salt, prodrug or derivative thereof 67. The method according to claim 61 characterized in that cell death occurs due to necrosis, apoptosis, or regulation of the trajectory. FAS 68. The method according to claim 61, characterized by or because the cell is hyperproliferative. 20 69. The method of compliance with the claim 61, characterized in that the cell or tissue is autoimmune or is affected by autoimmune reaction. 70. The method according to claim 61, characterized in that the cell or tissue is inflammatory or is affected by inflammation. 71. The method according to claim 61, characterized in that the cell is a monocytic cell. 72. The method according to claim 61, characterized in that the cell is infected with a virus. 73. The method according to claim 61, characterized in that it further comprises co-administering one or more additional agents to the cell. 74. The method according to claim 73, characterized in that the additional agent is selected from the group consisting of: chemotherapeutic agent, immunosuppressant, anti-inflammatory agent, antiviral agent, or radiation. 75. A method for improving the efficacy of an agent for treating an autoimmune disease characterized in that it comprises administering an effective amount of a benzodiazepine compound. 76. The method according to claim 75, characterized in that the benzodiazepine does not bind to a central benzodiazepine receptor and binds only with ______ ^ _____ | ___ Ég__ | _ | f ____ l ___ Í_É__! Y. low affinity to a peripheral benzodiazepine receptor. 77. The method according to claim 75 or 76, characterized in that the benzodiazepine induces apoptosis in a low serum assay. 78. The method according to claim 75, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein, R is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -Rs, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -Rβ-NH-C (= 0) - R7 or Rg-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl or heterocyclic; Y l_Í ___ í ___ | _llÉ _-___-__ l_______i______i Ák! ^ m i ^ í ^ tí ^ ^ k ^! i ** ^^ ** each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 79. The method according to claim 75, characterized in that the benzodiazepine is a compound having the structure: 10 or its enantiomer, wherein Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2 / -NHC (= 0) -R5 or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) - R7 or -Re-C (= 0) -NH-R7 wherein Rβ is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, ? ^^ - i__l -____ i __________? ______ i __ ^ M hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 80. The method according to claim 75, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -Rs, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) - R 15 or -R 6 -C (= 0) -NH-R- /, wherein R 6 is an aliphatic linker of 1-6 carbons and R 7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 81. A method for inhibiting viral proliferation in a virally infected cell characterized in that it comprises contacting the cell with an effective amount of one or more benzodiazepine compounds. 82. The method according to claim 81, characterized in that the benzodiazepine does not bind to a central benzodiazepine receptor and only binds with low affinity to a peripheral benzodiazepine receptor. F 83. The method according to claims 81 or 82, characterized in that benzodiazepine induces apoptosis in a low serum assay. 84. The method according to claim 15 81, characterized in that the benzodiazepine is a compound having the structure: «M ^ > _ _-_ É -_ --- _--- ^ -_-_- J_- «> . ^ * «- ~ * ~? * Mj Li ¿j aú? MA or its enantiomer, where Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) - R or R6-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl or heterocyclic; and each of R3 and R is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 85. The method according to claim 81, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2 -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) -R7 or -R6-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, 10 hydroxy, alkoxy, halo, amino, lower alkyl-amino F substituted, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 86. The method according to claim 81, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein, 20 Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2 / -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) - R or -R6-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 87. The method according to claim 81, characterized in that the viral infection is caused by a virus selected from the group consisting of herpes virus, hepatitis virus, or a retrovirus. 88. The method according to claim 87, characterized in that the virus is selected from the group consisting of HSV-1, HSV-2, HCMV, HBV, HCV, or HIV. 89. A method for identifying agents useful for treating a condition associated with a cell death process in a subject, characterized in that the method comprises contacting a cell maintained in low serum media with a test agent under conditions inducing death. cellular, and testing for cell death, so they identify useful agents to treat the l * UXLé * l * att * **. *, i > * - t -.- ^^ |? ^ ^ Condition associated with the process of cell death. 90. The method according to claim 89, characterized in that it further comprises determining whether the test agent binds to a benzodiazepine receptor 5 or binds with low affinity to a peripheral benzodiazepine receptor. 91. The method according to claim 89, characterized in that the cell death process is necrotic. 92. The method according to claim 89, characterized in that the cell death process is apoptotic. 93. The method according to claim 89, characterized in that the cell is selected from the group of 15 cells consisting of: autoimmune cell, inflammatory cell, hyperproliferative cell, virally infected cell, atherosclerotic cell or osteoarthritic cell. 94. The method according to claim 89, characterized in that the cell is a cell affected by 20 an autoimmune condition or a cell affected by an inflammatory condition. 95. The method according to claim 89, characterized in that it further comprises contacting a control cell maintained in low serum media with a 25 benzodiazepine compound under conditions that induce ^^ cell death. 96. The method according to claim 95, characterized in that the benzodiazepine compound has the structure: 5 or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) -R or -Rg-C (= 0) -NH-R7, wherein Rβ is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, lower alkyl-amino 15 substituted, acetylamino, hydroxyamino, an aliphatic group that it has 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 97. The method according to claim 95, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein 5 is aryl, heterocyclic, -R6-NH-C (= 0) -R7 or -R6-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; .. ^ A ^^ .. au ^^ Jj ^ í _ ^^ lt < > 1at.-__ *. li ^. or a pharmaceutically acceptable salt, prodrug or derivative thereof. 98. The method according to claim 75, characterized in that the benzodiazepine is a compound having the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -Rs, or a portion that participates in the formation of hydrogen bonding, wherein Rs is aryl, heterocyclic, -RS-NH-C (= 0) - R or -R6-C (= 0) -NH-R7, wherein Rg is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or ....., .... ~. ^ .. ta ¡Mrt¡a | M | BMaÉ, _ £ ___ .. derived from it. 99. The method according to claim 91, characterized in that the level of serum is less than or equal to about 10% by volume of the maintenance medium. 100. The method according to claim 91, characterized in that the serum level is less than or equal to about 5% by volume of the maintenance medium. 101. The method according to claim 91, characterized in that the level of serum is less than or equal to about 1% by volume of the maintenance medium. 102. The method according to claim 91, characterized in that the level of serum is less than, or equal to about 0.5% by volume of the maintenance medium. 103. The method according to claim 91, characterized in that the level of serum is less than or equal to about 0.2% by volume of the maintenance medium. 10 The method according to claim 95, characterized in that the benzodiazepine compound is detectably labeled. 105. The use of a benzodiazepine compound for treating a condition associated with dysregulation of the cell death process in a subject, characterized in that benzodiazepine is a compound having the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -Rs, or a portion that participates in the formation of hydrogen bonding, wherein Rs is aryl, heterocyclic, -R6-NH-C (= 0) - R7 • 10 or -R6-C (= 0) -NH-R7, wherein Rs is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group which 15 has 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 106. The use according to claim 105, characterized in that cell death is due to apoptosis necrosis or regulation of the FAS path. 107. The use according to claim 105, characterized in that the condition is an autoimmune disease selected from the group consisting of systemic lupus erythematosus, rheumatoid arthritis, Sjdgren's syndrome, graft-versus-host disease and myasthenia gravis. 108. The use according to claim 105, characterized in that the condition is a chronic inflammatory condition. 109. Use in accordance with the claim108, characterized in that the inflammatory condition is psoriasis, asthma, or Crohn's disease. 110. The use according to claim 105, characterized in that the condition is a hyperproliferative disorder or neoplasm. 111. The use according to claim 110, characterized in that the hyperproliferative disorder is selected from the group consisting of B-cell lymphoma, T-cell lymphoma, cancer, cancers with chemo-resistance, disorders related to deficient p53 expression, and j-_J-j-Í-i .- «- iiÉfa -, _-- M < ij ^ g and | M¡ ^ .- i - «^ - MiJ« > j - ^ - ^^ disorders related to overexpression of endogenous bcl-xL. 112. The use according to claim 105, characterized in that the condition is induced by a viral infection, wherein the viral infection is caused by a virus selected from the group consisting of herpes virus, papilloma virus and Human Immunodeficiency Virus. (HIV) 113. The use according to claim 105, characterized in that the condition is atherosclerosis or 10 osteoarthritis. F 114. A benzodiazepine compound, characterized because it has the structure: or its enantiomer, wherein, 15 Ri is aliphatic or aryl; blM *** »,. * ^ - * .... ^ I tM ^ MM ^^^^. ^ -_ ^ -_ ii |" R is aliphatic, aryl, -NH2, -NHC (= 0) -Rs , or a portion that participates in the formation of hydrogen bonding, wherein Rs is aryl, heterocyclic, -R6-NH-C (= 0) -R or R6-C (= 0) -NH-R7, wherein is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; 10 or a pharmaceutically acceptable salt, prodrug or • derived from it. 115. A benzodiazepine compound characterized because it has the structure: or its enantiomer, wherein Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2 / -NHC (= 0) -R, or a portion that participates in the formation of hydrogen bonding, ? hi _____ t.1t ^^^^^^^^^^^^ tum ^^^? ^^ i ^^^^^^ m ^ where R5 is aryl, heterocyclic, -Rß-NH-C (= 0) -R or -Rs-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1- ^ F6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. -áf 10 116. A benzodiazepine compound, characterized in that it has the structure: or its enantiomer, wherein, Ri is aliphatic or aryl; R2 is aliphatic, aryl, -NH2, -NHC (= 0) -R5, or a portion that participates in the formation of hydrogen bonding, wherein R5 is aryl, heterocyclic, -R6-NH-C (= 0) -R7 or -Rg-C (= 0) -NH-R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, 4fffÜ "~ '-Ta¿" -. «A ^ A ... ^. ^ A ^^, ^^ .. * ^^^^ hydroxy, alkoxy, halo, amino, lower alkyl-substituted amino, acetylamino , hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 117. A benzodiazepine compound characterized because it has the structure: or its enantiomer, wherein, R <10> is an optionally substituted bisphenyl; R2 is aliphatic, aryl, -NH2, or -NHC (= 0) -R5, wherein Rs is aplo, heterocyclic, -R6-NH-C (= 0) -R or -Rs-C (= 0) -NH -R7, wherein R6 is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, substituted lower alkyl-amino, acetylamino, hydroxyamino, an aliphatic group having 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 118. A benzodiazepine compound, characterized because it has the structure: or its enantiomer, • wherein, Ri is optionally substituted bisphenyl; R2 is aliphatic, aryl, -NH2, or -NHC (= 0) -R5, wherein Rs is aryl, heterocyclic, -R6-NH-C (= 0) -R 10 or -R5-C (= 0) -NH-R7, wherein Rg is an aliphatic linker of 1-6 carbons and R7 is aliphatic, aryl, or heterocyclic; and each of R3 and R4 is independently hydrogen, hydroxy, alkoxy, halo, amino, lower alkyl-amino F substituted, acylamino, hydroxyamino, an aliphatic group that 15 has 1-8 carbons and 1-20 hydrogens, aryl, or heterocyclic; or a pharmaceutically acceptable salt, prodrug or derivative thereof. 119. A compound characterized in that it has the structure: or its enantiomer, or a pharmaceutically acceptable salt, prodrug or derivative thereof. 120. A compound characterized in that it has the structure: or its enantiomer, or a pharmaceutically acceptable salt, prodrug or ísys-. - .. Ji: "*! Derived from it." 121. A compound characterized because it has the structure: or its enantiomer, or a pharmaceutically acceptable salt, prodrug or derivative thereof. 122. A compound characterized in that it has the structure: or its enantiomer, or a pharmaceutically acceptable salt, prodrug or derivative thereof. 123. A compound characterized in that it has the • structure: or its enantiomer, or a pharmaceutically acceptable salt, prodrug or derivative thereof. 124. A compound characterized in that it has the structure: iiÉifÉIBiriríf * ^ "" - "- -? -. * or its enantiomer, or a pharmaceutically acceptable salt, prodrug or derivative thereof. p 125. A compound characterized in that it has the 5 structure: or its enantiomer, or a pharmaceutically acceptable salt, prodrug or derivative thereof. 126. A compound characterized in that it has the 10 structure: or its enantiomer, or a pharmaceutically acceptable salt, prodrug or derivative thereof. • 127. The method according to claim 17, characterized in that the hyperproliferative disorder is neuroblastoma or ovarian cancer. 128. The method according to claim 49, characterized in that the hyperproliferative disorder is neuroblastoma or ovarian cancer. 129. The method according to claim 111, characterized in that the hyperproliferative disorder is neuroblastoma or ovarian cancer. * S ______) Benzodlazepine compounds and methods for using these compounds are provided. Some of the benzodiazepine compounds include the compounds 1,4- 5-benzodiazepine-2-one and 1,4-benzodiazepine-2,5-dione of the structures (I) or (II): wherein R_, R2 / R3 and R are as defined. The invention also includes enantiomers, pharmaceutically acceptable salts, prodrug or derivatives of the benzodiazepine compounds. Any one or more of these 10 benzodiazepine compounds can be used to treat • a variety of dysregulatory disorders related to cell death. Such disorders include autoimmune disorders, inflammatory conditions, hyperproliferative conditions, viral infections and arteriosclerosis. In addition, the above compounds can be used to prepare medicaments for treating the dysregulatory disorders described above. Benzodiazepines can also be used in drug selection and other tests • diagnostic methods. 20 01 / // osz
MXPA01011052A 1999-04-30 2000-04-27 Therapeutic applications of pro-apoptotic benzodiazepines. MXPA01011052A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US13176199P 1999-04-30 1999-04-30
US16551199P 1999-11-15 1999-11-15
US19185500P 2000-03-24 2000-03-24
PCT/US2000/011599 WO2000066106A2 (en) 1999-04-30 2000-04-27 Use of benzodiazepines for treating autoimmune diseases induced by apoptosis

Publications (1)

Publication Number Publication Date
MXPA01011052A true MXPA01011052A (en) 2002-11-22

Family

ID=27384197

Family Applications (1)

Application Number Title Priority Date Filing Date
MXPA01011052A MXPA01011052A (en) 1999-04-30 2000-04-27 Therapeutic applications of pro-apoptotic benzodiazepines.

Country Status (13)

Country Link
US (5) US7125866B1 (en)
EP (4) EP1143946B1 (en)
JP (1) JP5127096B2 (en)
AT (2) ATE258439T1 (en)
AU (2) AU782447C (en)
CA (1) CA2372150C (en)
DE (2) DE60007960T2 (en)
DK (1) DK1143946T3 (en)
ES (1) ES2214273T3 (en)
IL (2) IL146222A0 (en)
MX (1) MXPA01011052A (en)
PT (1) PT1143946E (en)
WO (1) WO2000066106A2 (en)

Families Citing this family (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2785803B1 (en) * 1998-11-17 2005-03-25 Sanofi Sa USE OF A SUBSTANCE BINDING TO THE PERIPHERAL RECEPTOR OF BENZODIAZEPINES IN THE TREATMENT OF SKIN STRESS
EP1143946B1 (en) * 1999-04-30 2004-01-28 The Regents Of The University Of Michigan Use of benzodiazepines for treating autoimmune diseases induced by apoptosis
US20040176358A1 (en) * 1999-04-30 2004-09-09 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
US20030119029A1 (en) * 1999-04-30 2003-06-26 Regents Of The University Of Michigan Compositions and methods relating to novel benzodiazepine compounds and targets thereof
US7572788B2 (en) * 1999-04-30 2009-08-11 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
US20060025388A1 (en) 1999-04-30 2006-02-02 Glick Gary D Compositions and methods relating to novel compounds and targets thereof
US7276348B2 (en) 1999-04-30 2007-10-02 Regents Of The University Of Michigan Compositions and methods relating to F1F0-ATPase inhibitors and targets thereof
US20050113460A1 (en) * 1999-04-30 2005-05-26 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
US7766013B2 (en) 2001-06-05 2010-08-03 Alexza Pharmaceuticals, Inc. Aerosol generating method and device
US20070122353A1 (en) 2001-05-24 2007-05-31 Hale Ron L Drug condensation aerosols and kits
AU2002332560B2 (en) * 2001-08-15 2006-01-19 The Regents Of The University Of Michigan Compositions and methods relating to novel benzodiazepine compounds and targets thereof
AU2002340464B2 (en) 2001-11-13 2008-03-13 3-Dimensional Pharmaceuticals, Inc. Substituted 1,4-benzodiazepines and uses thereof for the treatment of cancer
PL375860A1 (en) * 2002-09-20 2005-12-12 Arrow Therapeutics Limited Benzodiazepine derivatives and pharmaceutical compositions containing them
GB0221923D0 (en) * 2002-09-20 2002-10-30 Arrow Therapeutics Ltd Chemical compounds
WO2005004988A2 (en) * 2003-05-01 2005-01-20 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
WO2004100885A2 (en) * 2003-05-09 2004-11-25 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
US20050079166A1 (en) 2003-05-21 2005-04-14 Alexza Molecular Delivery Corporation Self-contained heating unit and drug-supply unit employing same
US20050120397A1 (en) * 2003-11-24 2005-06-02 Hermann Steller Compounds and methods for regulation of spermatid differentiation
WO2005090319A1 (en) * 2004-03-19 2005-09-29 Arrow Therapeutics Limited Process for preparing benzodiazepines
US20050272723A1 (en) * 2004-04-27 2005-12-08 The Regents Of The University Of Michigan Methods and compositions for treating diseases and conditions associated with mitochondrial function
US20060052369A1 (en) * 2004-09-07 2006-03-09 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
US7638624B2 (en) 2005-01-03 2009-12-29 The Regents Of The University Of Michigan Compositions and methods relating to novel benzodiazepine compounds and derivatives
JP2008545757A (en) * 2005-06-01 2008-12-18 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン Unsolvated benzodiazepine compositions and methods
US20090221559A1 (en) * 2005-09-02 2009-09-03 Jean-Francois Bonfanti Benzodiazepines as hcv inhibitors
WO2007034127A1 (en) * 2005-09-19 2007-03-29 Arrow Therapeutics Limited Benzodiazepine derivatives for treating hepatitis c infection
US20070105844A1 (en) * 2005-10-26 2007-05-10 Regents Of The University Of Michigan Therapeutic compositions and methods
EP2604269B1 (en) * 2005-11-01 2014-09-24 The Regents Of The University Of Michigan 1,4-benzodiazepine-2,5-diones with therapeutic properties
CN102516161A (en) 2006-03-29 2012-06-27 弗·哈夫曼-拉罗切有限公司 Pyridine and pyrimidine derivatives as mglur2 antagonists
US7759338B2 (en) * 2006-04-27 2010-07-20 The Regents Of The University Of Michigan Soluble 1,4 benzodiazepine compounds and stable salts thereof
CA2659549C (en) * 2006-06-09 2013-07-30 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
US20100021928A1 (en) * 2006-06-26 2010-01-28 Technion Research & Development Foundation Ltd. Methods and kits for diagnosing cancer
AR065374A1 (en) * 2007-02-16 2009-06-03 Tibotec Pharm Ltd 1, 1-DIOXO-1-TIA-5, 10-DIAZADIBENZOCICLOHEPTENOS USEFUL AS INHIBITORS OF THE VIRUSES OF HEPATITIS C, METHODS FOR THEIR PREPARATION, A PHARMACEUTICAL COMPOSITION THAT UNDERSTANDS AND ITS USE IN THE MANUFACTURE OF AN INHIBITION REPLACEMENT MEDICATION HCV
WO2008112553A1 (en) * 2007-03-09 2008-09-18 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
EP2121088B1 (en) 2007-03-09 2016-07-13 Alexza Pharmaceuticals, Inc. Heating unit for use in a drug delivery device
WO2008141081A1 (en) * 2007-05-10 2008-11-20 Amr Technology, Inc. Aryl- and heteroaryl-substituted tetrahydrobenzo-1,4-diazepines and use thereof to block reuptake of norepinephrine, dopamine, and serotonin
WO2009036175A2 (en) 2007-09-14 2009-03-19 The Regents Of The University Of Michigan F1f0-atpase inhibitors and related methods
CA2703005A1 (en) 2007-11-06 2009-05-14 The Regents Of The University Of Michigan Benzodiazepinone compounds useful in the treatment of skin conditions
GB2456562A (en) * 2008-01-18 2009-07-22 Petra Technology Ltd Treatment of HIV/AIDS
EP2145873A1 (en) * 2008-06-17 2010-01-20 Commissariat A L'energie Atomique New compounds with activity that protects against the action of toxins and viruses in intracellular action mode
JP5567573B2 (en) 2008-09-11 2014-08-06 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン Arylguanidine F1F0-ATPase inhibitors and methods related thereto
WO2010121164A2 (en) 2009-04-17 2010-10-21 The Regents Of The University Of Michigan 1,4-benzodiazepinone compounds and their use in treating cancer
US9075060B2 (en) 2009-06-16 2015-07-07 Technion Research & Development Foundation Limited Methods for diagnosing oral or oral-pharyngeal cancer
EP2470020A4 (en) * 2009-09-18 2013-03-13 Univ Michigan Benzodiazepinone compounds and methods of treatment using same
US8815845B2 (en) 2009-11-17 2014-08-26 The Regents Of The University Of Michigan 1,4-benzodiazepine-2,5-diones and related compounds with therapeutic properties
JP5856063B2 (en) 2009-11-17 2016-02-09 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン 1,4-Benzodiazepine-2,5-dione and related compounds having therapeutic properties
CN102190631B (en) * 2010-03-10 2014-10-29 中国人民解放军63975部队 Preparation method of benzodiazepine compound
TWI530489B (en) 2011-03-22 2016-04-21 必治妥美雅史谷比公司 Bis(fluoroalkyl)-1,4-benzodiazepinone compounds
CN104854094A (en) 2012-09-21 2015-08-19 百时美施贵宝公司 Fluoroalkyl dibenzodiazepinone compounds
TWI614238B (en) 2012-09-21 2018-02-11 必治妥美雅史谷比公司 Bis(fluoroalkyl)-1,4-benzodiazepinone compounds and prodrugs thereof
WO2014047393A1 (en) 2012-09-21 2014-03-27 Bristol-Myers Squibb Company N-substituted bis(fluoroalkyl)-1,4-benzodiazepinone compounds as notch inhibitors
CN104822677A (en) 2012-09-21 2015-08-05 百时美施贵宝公司 Fluoroalkyl-1,4-benzodiazepinone compounds
CN104797584A (en) 2012-09-21 2015-07-22 百时美施贵宝公司 Tricyclic heterocyclic compounds as notch inhibitors
EP2897944B1 (en) 2012-09-21 2016-10-26 Bristol-Myers Squibb Company Substituted 1,5-benzodiazepinone compounds
EP2897942B1 (en) 2012-09-21 2016-08-31 Bristol-Myers Squibb Company Fluoroalkyl and fluorocycloalkyl 1,4-benzodiazepinone compounds as notch inhibitors
US9273075B2 (en) 2012-09-21 2016-03-01 Bristol-Myers Squibb Company Prodrugs of 1,4-benzodiazepinone compounds
JP2015534554A (en) 2012-09-21 2015-12-03 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Alkyl, fluoroalkyl-1,4-benzodiazepinone compounds
CN105101968A (en) 2013-04-04 2015-11-25 百时美施贵宝公司 Combination therapy for the treatment of proliferative diseases
SI3277286T1 (en) 2015-03-31 2021-09-30 Enanta Pharmaceuticals, Inc. Bile acid derivatives as fxr/tgr5 agonists and methods of use thereof
US9957281B2 (en) 2015-07-22 2018-05-01 Enanta Pharmaceuticals, Inc. Benzodiazepine derivatives as RSV inhibitors
EP3402799B1 (en) 2016-01-15 2022-05-04 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as rsv inhibitors
CN105693634B (en) * 2016-03-17 2018-12-11 清华大学 Compound and application thereof
WO2017189663A1 (en) 2016-04-26 2017-11-02 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as fxr agonists and methods of use thereof
US10080741B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
WO2017189651A1 (en) 2016-04-26 2017-11-02 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as fxr agonists and methods of use thereof
US10450306B2 (en) 2016-10-04 2019-10-22 Enanta Pharmaceuticals, Inc. Isoxazole analogs as FXR agonists and methods of use thereof
WO2018081285A1 (en) 2016-10-26 2018-05-03 Enanta Pharmaceuticals, Inc. Urea-containing isoxazole derivatives as fxr agonists and methods of use thereof
US10398706B2 (en) 2017-01-06 2019-09-03 Enanta Pharmaceuticals, Inc. Heteroaryldiazepine derivatives as RSV inhibitors
BR112019016914A2 (en) 2017-02-16 2020-04-14 Enanta Pharm Inc processes for the preparation of benzodiazepine derivatives
US10752598B2 (en) 2017-06-07 2020-08-25 Enanta Pharmaceuticals, Inc. Aryldiazepine derivatives as RSV inhibitors
WO2019006295A1 (en) 2017-06-30 2019-01-03 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as rsv inhibitors
US10851115B2 (en) 2017-06-30 2020-12-01 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
AU2018339068B2 (en) 2017-09-29 2022-12-15 Enanta Pharmaceuticals, Inc. Combination pharmaceutical agents as RSV inhibitors
US10647711B2 (en) 2017-11-13 2020-05-12 Enanta Pharmaceuticals, Inc. Azepin-2-one derivatives as RSV inhibitors
AU2018365174B2 (en) 2017-11-13 2023-03-30 Enanta Pharmaceuticals, Inc. Processes for the resolution of benzodiazepin-2-one and benzoazepin-2-one derivatives
WO2019118571A1 (en) 2017-12-12 2019-06-20 Enanta Pharmaceuticals, Inc. Isoxazole analogs as fxr agonists and methods of use thereof
WO2019160813A1 (en) 2018-02-14 2019-08-22 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as fxr agonists and methods of use thereof
WO2019199908A1 (en) 2018-04-11 2019-10-17 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as rsv inhibitors
KR20210138684A (en) 2019-03-18 2021-11-19 이난타 파마슈티칼스, 인코포레이티드 Benzodiazepine derivatives as RSV inhibitors
US11179400B2 (en) 2019-04-09 2021-11-23 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
WO2020231917A1 (en) 2019-05-13 2020-11-19 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as fxr agonists and methods of use thereof
IL291834A (en) 2019-10-04 2022-06-01 Enanta Pharm Inc Antiviral heterocyclic compounds
US11505558B1 (en) 2019-10-04 2022-11-22 Enanta Pharmaceuticals, Inc. Antiviral heterocyclic compounds
UY39032A (en) 2020-01-24 2021-07-30 Enanta Pharm Inc HETEROCYCLIC COMPOUNDS AS ANTIVIRAL AGENTS
US11534439B2 (en) 2020-07-07 2022-12-27 Enanta Pharmaceuticals, Inc. Dihydroquinoxaline and dihydropyridopyrazine derivatives as RSV inhibitors
WO2022086840A1 (en) 2020-10-19 2022-04-28 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as anti-viral agents
UY39642A (en) 2021-02-26 2022-09-30 Enanta Pharm Inc ANTIVIRAL HETEROCYCLIC COMPOUNDS
CN117263873A (en) * 2022-06-15 2023-12-22 复旦大学 1, 4-benzodiazepine compounds and application thereof in preparation of antitumor drugs

Family Cites Families (110)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2457405A (en) * 1945-09-25 1948-12-28 Monsanto Chemicals Halo nitroparaffin modified aminoplasts
DE1810423U (en) 1960-01-05 1960-04-28 Kaiserslautern Guss Armatur CHIMNEY CLEANING PLUG.
US3374264A (en) * 1962-10-04 1968-03-19 Hoffmann La Roche N-haloacetyl-anthranilic acids and esters
US3261828A (en) 1962-10-04 1966-07-19 Hoffmann La Roche 3h-1,4-benzodiazepine-2,5(1h,4h)-dione compounds
FR1497456A (en) * 1964-06-15 1967-10-13 Clin Byla Ets Ortho-amino aryl ketimines, heterocyclic compounds attached to them and preparation of these various bodies
NO120269B (en) * 1965-11-02 1970-09-28 Hoffmann La Roche
US3415814A (en) 1966-09-28 1968-12-10 Sterling Drug Inc 4-(cyclopropylmethyl)-3h-1,4-benzodiazepine-2,5(1h,4h)-dione
US3384635A (en) 1966-09-28 1968-05-21 Sterling Drug Inc 1, 4-benzodiazepine derivatives
AT280290B (en) * 1967-11-24 1970-04-10 Boehringer Sohn Ingelheim Process for the preparation of new 1-phenyl-4-alkyl-3H-1,4-benzodiazepine-2,5- [1H, 4H] -diones
USRE30293E (en) * 1969-03-18 1980-06-03 Knoll A.G. Process for preparing 1,5-benzodiazepine-2-ones
US4108852A (en) * 1969-03-18 1978-08-22 Knoll Ag. Process for preparing 1,5-benzodiazepine-2-ones
CH559729A5 (en) 1970-10-23 1975-03-14 Hoechst Ag
GB1324469A (en) * 1970-10-28 1973-07-25 Knoll Ag Derivatives of benzodiazepines
US3806594A (en) * 1972-11-24 1974-04-23 Abbott Lab Method of reducing involuntary movements associated with cerebral palsy using clorazepate dipotassium
ZA7467B (en) * 1973-01-16 1974-11-27 J Voorhees Compositions and treatment of proliferative skin diseases with b2 agonists
IE38931B1 (en) 1973-03-09 1978-07-05 Lipha Triazolobenzodiazepines
US4110327A (en) * 1974-05-09 1978-08-29 Toyama Chemical Co., Ltd. 2,3 Diketo-piperazinocarbonylamino alkanoic acids and derivatives
US4076823A (en) 1977-08-18 1978-02-28 E. R. Squibb & Sons, Inc. Triazolo-2,4-benzodiazepines
JPS59112984A (en) * 1982-12-21 1984-06-29 Shionogi & Co Ltd 1,4-benzodiazepine derivative
US4495101A (en) 1983-04-28 1985-01-22 American Home Products Corporation Antiinflammatory 5H-tetrazolo (5,1-c)(1,4)benzodiazepine derivatives
US4551480A (en) * 1983-06-21 1985-11-05 Stiefel Laboratories, Inc. Compositions for the treatment of psoriasis
US4894366A (en) * 1984-12-03 1990-01-16 Fujisawa Pharmaceutical Company, Ltd. Tricyclo compounds, a process for their production and a pharmaceutical composition containing the same
US5004741A (en) * 1984-06-26 1991-04-02 Merck & Co., Inc. Methods of antagonizing CCK or gastrin with benzodiazepine analogs
US4820834A (en) * 1984-06-26 1989-04-11 Merck & Co., Inc. Benzodiazepine analogs
DE3435974A1 (en) * 1984-10-01 1986-04-10 Boehringer Ingelheim KG, 6507 Ingelheim PHARMACEUTICAL COMPOSITIONS CONTAINING DIAZEPINE WITH PAF-ANTAGONISTIC EFFECT
FR2591596B1 (en) 1985-12-13 1988-09-09 Roussel Uclaf NOVEL 4H-TRIAZOLO (4,3-A) (1,4) BENZODIAZEPINES, THEIR PREPARATION PROCESS, THEIR APPLICATION AS MEDICAMENTS AND THE COMPOSITIONS CONTAINING THEM
GB8608080D0 (en) * 1986-04-02 1986-05-08 Fujisawa Pharmaceutical Co Solid dispersion composition
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4751223A (en) 1986-10-14 1988-06-14 Hoechst-Roussel Pharmaceuticals, Inc. Antiinflammatory and analgesic aminoalkyl tetracyclic benzodiazepines
US4898861A (en) * 1987-03-26 1990-02-06 Hoffmann-La Roche Inc. Method for inhibiting the proliferation of tumor cells
US5147872A (en) * 1987-06-09 1992-09-15 Golwyn Daniel H Treatment of immunologically based disorders, specifically psoriasis
US4970207A (en) * 1988-07-07 1990-11-13 Fujisawa Pharmaceutical Company, Ltd. Benzodiazepine derivatives
WO1990013332A1 (en) 1989-05-11 1990-11-15 Cedars-Sinai Medical Center Stent with sustained drug delivery
US5258510A (en) * 1989-10-20 1993-11-02 Otsuka Pharma Co Ltd Benzoheterocyclic compounds
US5041438A (en) 1989-10-30 1991-08-20 Hoffmann-La Roche Inc. Method for treating retroviral infections with benzodiazepine compounds
US5324726A (en) 1989-12-18 1994-06-28 Merck & Co., Inc. Benzodiazepine analogs
EP0470246B1 (en) 1990-02-28 1995-06-28 Medtronic, Inc. Intralumenal drug eluting prosthesis
DE69123090D1 (en) * 1990-07-06 1996-12-19 Yoshitomi Pharmaceutical Condensed thiophene compounds and their use
GB9015879D0 (en) * 1990-07-19 1990-09-05 Fujisawa Pharmaceutical Co Benzodiazepine derivatives
BR9200951A (en) * 1991-03-21 1992-11-17 Hoffmann La Roche COMPOUNDS, PROCESS FOR ITS PRODUCTION, PHARMACEUTICAL PREPARATIONS AND USE
CA2130195A1 (en) 1992-03-16 1993-09-30 Mark Gary Bock Benzodiazepine derivatives, compositions containing them and their use in therapy
US5599352A (en) 1992-03-19 1997-02-04 Medtronic, Inc. Method of making a drug eluting stent
US5288514A (en) * 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
GB9307527D0 (en) * 1993-04-13 1993-06-02 Fujisawa Pharmaceutical Co New venzamide derivatives,processes for the preparation thereof and pharmaceutical composition comprising the same
US5391566A (en) * 1993-07-20 1995-02-21 Merck & Co., Inc. Benzimidazolinones substituted with phenoxyphenylacetic acid derivatives
US6524832B1 (en) * 1994-02-04 2003-02-25 Arch Development Corporation DNA damaging agents in combination with tyrosine kinase inhibitors
US5444092A (en) * 1994-07-20 1995-08-22 Collins; Jerry Method and composition for treating psoriasis
US5763437A (en) * 1994-07-29 1998-06-09 Fujisawa Pharmaceutical Co., Ltd. Benzodiazepine derivatives
EP0776203A4 (en) * 1994-08-18 1997-12-29 Merck & Co Inc N-2,3-dihydro-1-(2-propyl)-2-oxo-5-phenyl-1h-1,4-benzodiazepines
US5618792A (en) 1994-11-21 1997-04-08 Cortech, Inc. Substituted heterocyclic compounds useful as inhibitors of (serine proteases) human neutrophil elastase
AU5752696A (en) 1995-05-18 1996-11-29 Regents Of The University Of Michigan, The Dna binding antibodies
US5692337A (en) * 1995-06-07 1997-12-02 Motz, Jr.; Ronald W. Collapsible plant shelter
US5677282A (en) * 1995-06-07 1997-10-14 Proscript, Inc. Amino acid amides of 1,3,4-thiadiazoles as matrix metalloproteinase
US5962337A (en) 1995-06-29 1999-10-05 Pharmacopeia, Inc. Combinatorial 1,4-benzodiazepin-2,5-dione library
RU2096044C1 (en) 1995-07-19 1997-11-20 Малое предприятие "Ветта" Veterinary implantable preparation for regulation of biological rhythm in animals
US5776946A (en) 1995-08-28 1998-07-07 Mcgeer; Patrick L. Peripheral benzodiazepine receptor ligands as antiinflammatory agents
WO1997007828A1 (en) 1995-08-30 1997-03-06 The Regents Of The University Of California Therapy for cellular accumulation in chronic inflammatory diseases
ATE214055T1 (en) * 1995-09-21 2002-03-15 Inst Med Molecular Design Inc RETINOID POTENTIFIING COMPOUNDS
US6579854B1 (en) * 1996-08-14 2003-06-17 Vanderbilt University Diagnosis and management of infection caused by chlamydia
UA60311C2 (en) * 1996-10-02 2003-10-15 Смітклайн Бічам Корпорейшн Vitronectin receptor antagonists
US7351421B2 (en) * 1996-11-05 2008-04-01 Hsing-Wen Sung Drug-eluting stent having collagen drug carrier chemically treated with genipin
WO1998025911A1 (en) * 1996-12-10 1998-06-18 Zeria Pharmaceutical Co., Ltd. 1,5-benzodiazepine derivatives
US6074859A (en) * 1997-07-08 2000-06-13 Kikkoman Corporation Mutant-type bioluminescent protein, and process for producing the mutant-type bioluminescent protein
ES2256959T3 (en) * 1997-10-08 2006-07-16 Isotechnika,Inc. DEUTERED AND NON-DEUTERATED CYCLOSPORINE ANALOGS AND ITS USE AS IMMUNOMODULATING AGENTS.
US6100254A (en) * 1997-10-10 2000-08-08 Board Of Regents, The University Of Texas System Inhibitors of protein tyrosine kinases
FR2772271B1 (en) * 1997-12-11 2000-09-01 Union Pharma Scient Appl NEW PHARMACEUTICAL ASSOCIATION WITH ANALGESIC ACTIVITY
US6206914B1 (en) * 1998-04-30 2001-03-27 Medtronic, Inc. Implantable system with drug-eluting cells for on-demand local drug delivery
WO1999058117A1 (en) * 1998-05-13 1999-11-18 Sanofi-Synthelabo Use of compounds for reducing apoptosis
FR2779963A1 (en) 1998-06-22 1999-12-24 Centre Nat Rech Scient New composition useful for treat cancer, comprises compound with affinity for mitochondrial benzodiazepine receptors and agent to induce apoptosis
US6277844B1 (en) 1998-09-14 2001-08-21 Sydney Spector Compound for selective treatment of malignant cells by inhibiting cell cycle progression, decreasing Bcl2, and increasing apoptosis
US20030044776A1 (en) * 1998-09-25 2003-03-06 James A. Dykens Compositions and methods for identifying agents that alter mitochondrial permeability transition pores
FR2785803B1 (en) 1998-11-17 2005-03-25 Sanofi Sa USE OF A SUBSTANCE BINDING TO THE PERIPHERAL RECEPTOR OF BENZODIAZEPINES IN THE TREATMENT OF SKIN STRESS
US7175953B2 (en) * 1999-04-09 2007-02-13 Institute Fuer Diagnostik Forschung Short-warp peptide-dye conjugate as contrast agent for optical diagnostic
EP1143946B1 (en) * 1999-04-30 2004-01-28 The Regents Of The University Of Michigan Use of benzodiazepines for treating autoimmune diseases induced by apoptosis
US20030119029A1 (en) * 1999-04-30 2003-06-26 Regents Of The University Of Michigan Compositions and methods relating to novel benzodiazepine compounds and targets thereof
US20040176358A1 (en) * 1999-04-30 2004-09-09 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
US7276348B2 (en) * 1999-04-30 2007-10-02 Regents Of The University Of Michigan Compositions and methods relating to F1F0-ATPase inhibitors and targets thereof
US7572788B2 (en) * 1999-04-30 2009-08-11 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
US20060025388A1 (en) * 1999-04-30 2006-02-02 Glick Gary D Compositions and methods relating to novel compounds and targets thereof
US20050113460A1 (en) 1999-04-30 2005-05-26 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
AU5826100A (en) 1999-07-13 2001-01-30 F. Hoffmann-La Roche Ag Benzazepinones and quinazolines
US6524623B1 (en) * 1999-11-12 2003-02-25 Milton Hodosh Therapeutic compositions and methods of use thereof
WO2001085218A2 (en) * 2000-05-11 2001-11-15 Eastman Chemical Company Acylated cyclodextrin guest inclusion complexes
US20020048566A1 (en) * 2000-09-14 2002-04-25 El-Deiry Wafik S. Modulation of cellular apoptosis and methods for treating cancer
CA2432825A1 (en) 2000-12-15 2002-06-20 Emory University Nonpeptide agonists and antagonists of vasopressin receptors
EP1392663A2 (en) * 2001-06-07 2004-03-03 Neuro3D Cyclic nucleotide phosphodiesterase inhibitors, preparation and uses thereof
AU2002332560B2 (en) 2001-08-15 2006-01-19 The Regents Of The University Of Michigan Compositions and methods relating to novel benzodiazepine compounds and targets thereof
WO2003050261A2 (en) * 2001-12-10 2003-06-19 Bristol-Myers Squibb Company (1-phenyl-2-heteroaryl)ethyl-guanidine compounds as inhibitors of mitochondrial f1f0 atp hydrolase
US20040009972A1 (en) * 2002-06-17 2004-01-15 Ding Charles Z. Benzodiazepine inhibitors of mitochondial F1F0 ATP hydrolase and methods of inhibiting F1F0 ATP hydrolase
US7150433B2 (en) * 2002-08-16 2006-12-19 Toyota Motor Sales, U.S.A., Inc. Aircraft bolster trays
US20040087489A1 (en) * 2002-11-06 2004-05-06 Antonio Ruiz Compositions and methods for the treatment of mycobacterial infections
AU2003295402A1 (en) * 2002-11-06 2004-06-03 Bristol-Myers Squibb Company Acyl guanidine compounds and use thereof
CA2508542A1 (en) * 2002-12-03 2004-06-17 Vela Pharmaceuticals, Inc. Pharmaceutical composition of 1-(3,4-dimethoxyphenyl)-4-methyl-5-ethyl-7-methoxy-8-hydroxy-5h-2,3-benzodiazepine and uses thereof
WO2004050610A2 (en) 2002-12-04 2004-06-17 Gene Logic Inc. Modulators of melanocortin receptor
WO2005004988A2 (en) 2003-05-01 2005-01-20 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
US7351241B2 (en) * 2003-06-02 2008-04-01 Carl Zeiss Meditec Ag Method and apparatus for precision working of material
US20090275099A1 (en) 2004-04-27 2009-11-05 Regents Of The University Of Michigan Methods and compositions for treating diseases and conditions associated with mitochondrial function
US20050272723A1 (en) 2004-04-27 2005-12-08 The Regents Of The University Of Michigan Methods and compositions for treating diseases and conditions associated with mitochondrial function
AU2005262322B2 (en) 2004-07-01 2012-07-26 Synta Pharmaceuticals Corp. 2-substituted heteroaryl compounds
US20060052369A1 (en) * 2004-09-07 2006-03-09 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
US7638624B2 (en) * 2005-01-03 2009-12-29 The Regents Of The University Of Michigan Compositions and methods relating to novel benzodiazepine compounds and derivatives
JP2008545757A (en) 2005-06-01 2008-12-18 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン Unsolvated benzodiazepine compositions and methods
AU2006306347A1 (en) 2005-10-26 2007-05-03 The Regents Of The University Of Michigan Therapeutic compositions and methods
US20070105844A1 (en) * 2005-10-26 2007-05-10 Regents Of The University Of Michigan Therapeutic compositions and methods
EP2604269B1 (en) 2005-11-01 2014-09-24 The Regents Of The University Of Michigan 1,4-benzodiazepine-2,5-diones with therapeutic properties
US7759338B2 (en) * 2006-04-27 2010-07-20 The Regents Of The University Of Michigan Soluble 1,4 benzodiazepine compounds and stable salts thereof
CA2659549C (en) 2006-06-09 2013-07-30 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
WO2008112553A1 (en) * 2007-03-09 2008-09-18 The Regents Of The University Of Michigan Compositions and methods relating to novel compounds and targets thereof
WO2009036175A2 (en) 2007-09-14 2009-03-19 The Regents Of The University Of Michigan F1f0-atpase inhibitors and related methods
CA2703005A1 (en) 2007-11-06 2009-05-14 The Regents Of The University Of Michigan Benzodiazepinone compounds useful in the treatment of skin conditions

Also Published As

Publication number Publication date
US7125866B1 (en) 2006-10-24
WO2000066106A3 (en) 2001-08-16
WO2000066106A2 (en) 2000-11-09
DE60007960D1 (en) 2004-03-04
EP2359831A2 (en) 2011-08-24
IL146222A0 (en) 2002-07-25
ATE258439T1 (en) 2004-02-15
US8809323B2 (en) 2014-08-19
DE60007960T2 (en) 2004-10-21
WO2000066106A9 (en) 2002-04-18
EP1398033B1 (en) 2009-06-17
AU2005227385B2 (en) 2008-12-11
EP1398033A2 (en) 2004-03-17
PT1143946E (en) 2004-05-31
US20070036854A1 (en) 2007-02-15
EP2062583A3 (en) 2009-12-30
DE60042425D1 (en) 2009-07-30
EP2062583B1 (en) 2012-12-26
US20010016583A1 (en) 2001-08-23
AU4680200A (en) 2000-11-17
CA2372150A1 (en) 2000-11-09
AU2005227385C1 (en) 2009-05-21
AU2005227385A1 (en) 2005-12-01
US20140038947A1 (en) 2014-02-06
DK1143946T3 (en) 2004-06-07
CA2372150C (en) 2011-08-30
AU782447C (en) 2006-07-13
JP2002543121A (en) 2002-12-17
AU782447B2 (en) 2005-07-28
EP2359831A3 (en) 2012-02-01
IL146222A (en) 2008-06-05
EP2062583A2 (en) 2009-05-27
ATE433752T1 (en) 2009-07-15
US7220739B2 (en) 2007-05-22
JP5127096B2 (en) 2013-01-23
EP1143946B1 (en) 2004-01-28
EP1398033A3 (en) 2004-06-30
US20090012065A1 (en) 2009-01-08
ES2214273T3 (en) 2004-09-16
EP1143946A2 (en) 2001-10-17

Similar Documents

Publication Publication Date Title
MXPA01011052A (en) Therapeutic applications of pro-apoptotic benzodiazepines.
US20030119029A1 (en) Compositions and methods relating to novel benzodiazepine compounds and targets thereof
US8722667B2 (en) Compositions and methods for inhibiting the proliferation of cells
AU2006308655B2 (en) Novel 1,4-benzodiazepine-2,5-diones with therapeutic properties
AU2002332560B2 (en) Compositions and methods relating to novel benzodiazepine compounds and targets thereof
AU2002332560A1 (en) Compositions and methods relating to novel benzodiazepine compounds and targets thereof
MXPA05011741A (en) Compositions and methods relating to novel compounds and targets thereof.
AU2006201605B2 (en) Compositions and methods relating to novel benzodiazepine compounds and targets thereof

Legal Events

Date Code Title Description
FG Grant or registration